WO2023102463A1 - Procédés d'utilisation d'agents d'interleukine-2 - Google Patents

Procédés d'utilisation d'agents d'interleukine-2 Download PDF

Info

Publication number
WO2023102463A1
WO2023102463A1 PCT/US2022/080728 US2022080728W WO2023102463A1 WO 2023102463 A1 WO2023102463 A1 WO 2023102463A1 US 2022080728 W US2022080728 W US 2022080728W WO 2023102463 A1 WO2023102463 A1 WO 2023102463A1
Authority
WO
WIPO (PCT)
Prior art keywords
fold
seq
amino acid
fusion protein
variant
Prior art date
Application number
PCT/US2022/080728
Other languages
English (en)
Inventor
Gregory Babcock
Wayne Hancock
Original Assignee
Visterra, Inc.
The Children's Hospital Of Philadelphia
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Visterra, Inc., The Children's Hospital Of Philadelphia filed Critical Visterra, Inc.
Publication of WO2023102463A1 publication Critical patent/WO2023102463A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2013IL-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/436Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having oxygen as a ring hetero atom, e.g. rapamycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection

Definitions

  • Interleukin-2 is a cytokine that regulates the activities of the immune system. It is produced by leukocytes, such as T cells, natural killer (NK) cells, dendritic cells, and macrophages, in response to antigenic or mitogenic stimulation. IL-2 is important for T cell proliferation, B cell stimulation, and other activities associated with immunity and tolerance. It is part of the body’s adaptive immune response and discriminates between foreign and host antigens. IL-2 mediates its effects by binding to IL-2 receptors, which in turn activate downstream signaling events.
  • leukocytes such as T cells, natural killer (NK) cells, dendritic cells, and macrophages
  • IL-2 is important for T cell proliferation, B cell stimulation, and other activities associated with immunity and tolerance. It is part of the body’s adaptive immune response and discriminates between foreign and host antigens. IL-2 mediates its effects by binding to IL-2 receptors, which in turn activate downstream signaling events.
  • Human IL-2 is an-FDA approved drug for the treatment of diseases such as metastatic renal carcinoma and melanoma.
  • the use of IL-2 in eligible patients is sometimes restricted due to the severe toxicity associated with IL-2 therapy, and only a small subset of eligible patients will actually receive therapy.
  • the toxicities associated with IL-2 therapy can include severe fever, nausea, vomiting, vascular leak and serious hypotension. Despite these toxicities, however, IL-2 is typically effective for its approved indications.
  • the disclosure provides a method of treating a transplant rejection or a symptom thereof, comprising administering to a subject in need thereof an effective amount of an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, thereby treating the transplant rejection or the symptom thereof.
  • an IL-2 agent described herein e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein
  • the transplant is a heart transplant.
  • the level of Tregs in the subject is increased by about 2, 3, 4, 5, 6, 7, 8, 9, 10-fold or more, compared to the level of Tregs in the subject prior to administration of the IL-2 fusion protein. In an embodiment, the level of Tregs is determined in a sample from the subject.
  • the IL-2 fusion protein comprises an IL-2 variant described herein.
  • the IL-2 variant comprising: (i) (a) the amino acid substitution H16L or H16N, (b) the amino acid substitution I92S, or (c) both (a) and (b); and (ii) the amino acid substitutions V69A, Q74P, and C125S, corresponding to human IL-2 (SEQ ID NO: 1031).
  • the IL-2 variant further comprises the amino acid substitution T3A.
  • the IL-2 variant comprises the amino acid sequence of any of SEQ ID NOs: 4, 5, 11, 1000, 1001, or 1002, an amino acid sequence that is at least 95% identical thereto or differs by no more than 1, 2, 3, 4, or 5 amino acids therefrom, or a functional fragment thereof.
  • the IL-2 fusion protein further comprises an Fc region.
  • the Fc region comprises an Fc region of IgGl allotype m3 comprising an N297G substitution according to EU numbering.
  • the Fc region comprises the amino acid sequence of SEQ ID NO: 1003, or an amino acid sequence that is at least 95% identical thereto or differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids therefrom, or a functional fragment thereof.
  • the Fc region is fused to the C-terminus of the IL-2 variant.
  • the IL-2 fusion protein further comprises a linker.
  • the linker comprises IG+SA (SEQ ID NO: 48).
  • the fusion protein forms a dimer.
  • the fusion protein comprises an amino acid sequence of any of SEQ ID NOs: 1004, 1005, 1006, 1007, 1008, or 1009, an amino acid sequence that is at least 95% identical thereto or differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids therefrom, or a functional fragment thereof.
  • the method further comprises administering an immunosuppressive agent to the subject.
  • the immunosuppressive agent comprises rapamycin.
  • the subject is a human or a non-human primate. In an embodiment, the subject is a mouse.
  • the disclosure provides a method of preventing a transplant rejection or a symptom thereof, comprising administering to a subject in need thereof an effective amount of an IL- 2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, thereby preventing the transplant rejection or the symptom thereof.
  • an IL- 2 agent described herein e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein
  • the transplant is a heart transplant.
  • the level of Tregs in the subject is increased by about 2, 3, 4, 5, 6, 7, 8, 9, 10-fold or more, compared to the level of Tregs in the subject prior to administration of the IL-2 fusion protein. In an embodiment, the level of Tregs is determined in a sample from the subject.
  • the IL-2 fusion protein comprises an IL-2 variant described herein.
  • the IL-2 variant comprising: (i) (a) the amino acid substitution H16L or H16N, (b) the amino acid substitution I92S, or (c) both (a) and (b); and (ii) the amino acid substitutions V69A, Q74P, and C125S, corresponding to human IL-2 (SEQ ID NO: 1031).
  • the IL-2 variant further comprises the amino acid substitution T3A.
  • the IL-2 variant comprises the amino acid sequence of any of SEQ ID NOs: 4, 5, 11, 1000, 1001, or 1002, an amino acid sequence that is at least 95% identical thereto or differs by no more than 1, 2, 3, 4, or 5 amino acids therefrom, or a functional fragment thereof.
  • the IL-2 fusion protein further comprises an Fc region.
  • the Fc region comprises an Fc region of IgGl allotype m3 comprising an N297G substitution according to EU numbering.
  • the Fc region comprises the amino acid sequence of SEQ ID NO: 1003, or an amino acid sequence that is at least 95% identical thereto or differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids therefrom, or a functional fragment thereof.
  • the Fc region is fused to the C-terminus of the IL-2 variant.
  • the IL-2 fusion protein further comprises a linker.
  • the linker comprises (G+S)4 (SEQ ID NO: 48).
  • the fusion protein forms a dimer.
  • the fusion protein comprises an amino acid sequence of any of SEQ ID NOs: 1004, 1005, 1006, 1007, 1008, or 1009, an amino acid sequence that is at least 95% identical thereto or differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids therefrom, or a functional fragment thereof.
  • the method further comprises administering an immunosuppressive agent to the subject.
  • the immunosuppressive agent comprises rapamycin.
  • the subject is a human or a non-human primate. In an embodiment, the subject is a mouse.
  • the disclosure provides a method of modulating (e.g., increasing or inducing) immunosuppression for a transplant, comprising administering to a subject in need thereof an effective amount of an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL- 2 fusion protein described herein, thereby modulating (e.g., increasing or inducing) immunosuppression for a transplant.
  • an IL-2 agent described herein e.g., an IL-2 variant described herein, or an IL- 2 fusion protein described herein
  • the subject has received, is receiving, or will receive a transplant, when the IL-2 agent is administered to the subject.
  • the transplant is a heart transplant.
  • the level of Tregs in the subject is increased by about 2, 3, 4, 5, 6, 7, 8, 9, 10-fold or more, compared to the level of Tregs in the subject prior to administration of the IL-2 fusion protein. In an embodiment, the level of Tregs is determined in a sample from the subject.
  • the IL-2 fusion protein comprises an IL-2 variant described herein.
  • the IL-2 variant comprising: (i) (a) the amino acid substitution H16L or H16N, (b) the amino acid substitution I92S, or (c) both (a) and (b); and (ii) the amino acid substitutions V69A, Q74P, and C125S, corresponding to human IL-2 (SEQ ID NO: 1031).
  • the IL-2 variant further comprises the amino acid substitution T3A.
  • the IL-2 variant comprises the amino acid sequence of any of SEQ ID NOs: 4, 5, 11, 1000, 1001, or 1002, an amino acid sequence that is at least 95% identical thereto or differs by no more than 1, 2, 3, 4, or 5 amino acids therefrom, or a functional fragment thereof.
  • the IL-2 fusion protein further comprises an Fc region.
  • the Fc region comprises an Fc region of IgGl allotype m3 comprising an N297G substitution according to EU numbering.
  • the Fc region comprises the amino acid sequence of SEQ ID NO: 1003, or an amino acid sequence that is at least 95% identical thereto or differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids therefrom, or a functional fragment thereof.
  • the Fc region is fused to the C-terminus of the IL-2 variant.
  • the IL-2 fusion protein further comprises a linker.
  • the linker comprises (648)4 (SEQ ID NO: 48).
  • the fusion protein forms a dimer.
  • the fusion protein comprises an amino acid sequence of any of SEQ ID NOs: 1004, 1005, 1006, 1007, 1008, or 1009, an amino acid sequence that is at least 95% identical thereto or differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids therefrom, or a functional fragment thereof.
  • the method further comprises administering an immunosuppressive agent to the subject.
  • the immunosuppressive agent comprises rapamycin.
  • the subject is a human or a non-human primate. In an embodiment, the subject is a mouse.
  • the disclosure provides a method of conditioning a subject prior to a transplant, comprising administering to a subject in need thereof an effective amount of an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, thereby conditioning the subject prior to the transplant.
  • an IL-2 agent described herein e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein
  • the transplant is a heart transplant.
  • the level of Tregs in the subject is increased by about 2, 3, 4, 5, 6, 7, 8, 9, 10-fold or more, compared to the level of Tregs in the subject prior to administration of the IL-2 fusion protein. In an embodiment, the level of Tregs is determined in a sample from the subject.
  • the IL-2 fusion protein comprises an IL-2 variant described herein.
  • the IL-2 variant comprising: (i) (a) the amino acid substitution H16L or H16N, (b) the amino acid substitution I92S, or (c) both (a) and (b); and (ii) the amino acid substitutions V69A, Q74P, and C125S, corresponding to human IL-2 (SEQ ID NO: 1031).
  • the IL-2 variant further comprises the amino acid substitution T3A.
  • the IL-2 variant comprises the amino acid sequence of any of SEQ ID NOs: 4, 5, 11, 1000, 1001, or 1002, an amino acid sequence that is at least 95% identical thereto or differs by no more than 1, 2, 3, 4, or 5 amino acids therefrom, or a functional fragment thereof.
  • the IL-2 fusion protein further comprises an Fc region.
  • the Fc region comprises an Fc region of IgGl allotype m3 comprising an N297G substitution according to EU numbering.
  • the Fc region comprises the amino acid sequence of SEQ ID NO: 1003, or an amino acid sequence that is at least 95% identical thereto or differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids therefrom, or a functional fragment thereof.
  • the Fc region is fused to the C-terminus of the IL-2 variant.
  • the IL-2 fusion protein further comprises a linker.
  • the linker comprises (648)4 (SEQ ID NO: 48).
  • the fusion protein forms a dimer.
  • the fusion protein comprises an amino acid sequence of any of SEQ ID NOs: 1004, 1005, 1006, 1007, 1008, or 1009, an amino acid sequence that is at least 95% identical thereto or differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids therefrom, or a functional fragment thereof.
  • the method further comprises administering an immunosuppressive agent to the subject.
  • the immunosuppressive agent comprises rapamycin.
  • the subject is a human or a non-human primate. In an embodiment, the subject is a mouse.
  • the disclosure provides a method of selectively increasing Tregs, comprising administering to a subject in need thereof an effective amount of an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, wherein the subject has received, is receiving, or will receive a transplant, thereby selectively increasing Tregs.
  • an IL-2 agent described herein e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein
  • the transplant is a heart transplant.
  • the level of Tregs in the subject is increased by about 2, 3, 4, 5, 6, 7, 8, 9, 10-fold or more, compared to the level of Tregs in the subject prior to administration of the IL-2 fusion protein. In an embodiment, the level of Tregs is determined in a sample from the subject.
  • the IL-2 fusion protein comprises an IL-2 variant described herein.
  • the IL-2 variant comprising: (i) (a) the amino acid substitution H16L or H16N, (b) the amino acid substitution I92S, or (c) both (a) and (b); and (ii) the amino acid substitutions V69A, Q74P, and C125S, corresponding to human IL-2 (SEQ ID NO: 1031).
  • the IL-2 variant further comprises the amino acid substitution T3A.
  • the IL-2 variant comprises the amino acid sequence of any of SEQ ID NOs: 4, 5, 11, 1000, 1001, or 1002, an amino acid sequence that is at least 95% identical thereto or differs by no more than 1, 2, 3, 4, or 5 amino acids therefrom, or a functional fragment thereof.
  • the IL-2 fusion protein further comprises an Fc region.
  • the Fc region comprises an Fc region of IgGl allotype m3 comprising an N297G substitution according to EU numbering.
  • the Fc region comprises the amino acid sequence of SEQ ID NO: 1003, or an amino acid sequence that is at least 95% identical thereto or differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids therefrom, or a functional fragment thereof.
  • the Fc region is fused to the C-terminus of the IL-2 variant.
  • the IL-2 fusion protein further comprises a linker.
  • the linker comprises (648)4 (SEQ ID NO: 48).
  • the fusion protein forms a dimer.
  • the fusion protein comprises an amino acid sequence of any of SEQ ID NOs: 1004, 1005, 1006, 1007, 1008, or 1009, an amino acid sequence that is at least 95% identical thereto or differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids therefrom, or a functional fragment thereof.
  • the method further comprises administering an immunosuppressive agent to the subject.
  • the immunosuppressive agent comprises rapamycin.
  • the subject is a human or a non-human primate. In an embodiment, the subject is a mouse.
  • the disclosure provides a method of treating a heart disease or a symptom thereof, comprising administering to a subject in need thereof an effective amount of an IL- 2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein; and providing a heart transplant to the subject, thereby treating the heart disease or the symptom thereof.
  • an IL- 2 agent described herein e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein
  • the heart transplant is provided to the subject prior to, concurrent with, or after administration of the IL-2 fusion protein.
  • the level of Tregs in the subject is increased by about 2, 3, 4, 5, 6, 7, 8, 9, 10-fold or more, compared to the level of Tregs in the subject prior to administration of the IL-2 fusion protein. In an embodiment, the level of Tregs is determined in a sample from the subject.
  • the IL-2 fusion protein comprises an IL-2 variant described herein.
  • the IL-2 variant comprising: (i) (a) the amino acid substitution H16L or H16N, (b) the amino acid substitution I92S, or (c) both (a) and (b); and (ii) the amino acid substitutions V69A, Q74P, and C125S, corresponding to human IL-2 (SEQ ID NO: 1031).
  • the IL-2 variant further comprises the amino acid substitution T3A.
  • the IL-2 variant comprises the amino acid sequence of any of SEQ ID NOs: 4, 5, 11, 1000, 1001, or 1002, an amino acid sequence that is at least 95% identical thereto or differs by no more than 1, 2, 3, 4, or 5 amino acids therefrom, or a functional fragment thereof.
  • the IL-2 fusion protein further comprises an Fc region.
  • the Fc region comprises an Fc region of IgGl allotype m3 comprising an N297G substitution according to EU numbering.
  • the Fc region comprises the amino acid sequence of SEQ ID NO: 1003, or an amino acid sequence that is at least 95% identical thereto or differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids therefrom, or a functional fragment thereof.
  • the Fc region is fused to the C-terminus of the IL-2 variant.
  • the IL-2 fusion protein further comprises a linker.
  • the linker comprises IG+SL (SEQ ID NO: 48).
  • the fusion protein forms a dimer.
  • the fusion protein comprises an amino acid sequence of any of SEQ ID NOs: 1004, 1005, 1006, 1007, 1008, or 1009, an amino acid sequence that is at least 95% identical thereto or differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids therefrom, or a functional fragment thereof.
  • the method further comprises administering an immunosuppressive agent to the subject.
  • the immunosuppressive agent comprises rapamycin.
  • the subject is a human or a non-human primate. In an embodiment, the subject is a mouse.
  • the disclosure provides a combination comprising an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, and a transplant.
  • the transplant is a heart transplant.
  • the transplant further comprises one or more additional immunosuppressive agents, e.g., rapamycin.
  • the combination is for use in treating or preventing a transplant rejection in a subject.
  • the transplant rejection is a heart transplant rejection.
  • the IL-2 agent is administered to the subject prior to, during, and/or after the transplant.
  • the combination is for use in modulating (e.g., increasing or inducing), immunosuppression in a subject.
  • the combination is for use in selectively increasing T regulatory cells in a subject.
  • the subject is a human or a non-human primate.
  • the subject is a mouse.
  • the disclosure provides an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, for use in a method of treating a transplant rejection or a symptom thereof in a subject, as described herein.
  • an IL-2 agent described herein e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, for use in a method of treating a transplant rejection or a symptom thereof in a subject, as described herein.
  • the disclosure provides an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, for use in a method of preventing a transplant rejection or a symptom thereof in a subject, as described herein.
  • an IL-2 agent described herein e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, for use in a method of preventing a transplant rejection or a symptom thereof in a subject, as described herein.
  • the disclosure provides an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, for use in a method of modulating (e.g., increasing or inducing) immunosuppression for a transplant in a subject, as described herein.
  • an IL-2 agent described herein e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, for use in a method of modulating (e.g., increasing or inducing) immunosuppression for a transplant in a subject, as described herein.
  • the disclosure provides an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, for use in a method of conditioning a subject prior to a transplant, as described herein.
  • an IL-2 agent described herein e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, for use in a method of conditioning a subject prior to a transplant, as described herein.
  • the disclosure provides an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, for use in a method of selectively increasing Tregs in a subject, as described herein, e.g., wherein the subject has received, is receiving, or will receive a transplant.
  • an IL-2 agent described herein e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein
  • the disclosure provides an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, in combination with a heart transplant, for use in a method of treating a heart disease or a symptom thereof in a subject, as described herein.
  • the disclosure provides use of an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, in the manufacture of a medicament for treating a transplant rejection or a symptom thereof in a subject, as described herein.
  • the disclosure provides use of an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, in the manufacture of a medicament for preventing a transplant rejection or a symptom thereof in a subject, as described herein.
  • an IL-2 agent described herein e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, in the manufacture of a medicament for preventing a transplant rejection or a symptom thereof in a subject, as described herein.
  • the disclosure provides use of an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, in the manufacture of a medicament for modulating (e.g., increasing or inducing) immunosuppression for a transplant in a subject, as described herein.
  • an IL-2 agent described herein e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein
  • the disclosure provides use of an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, in the manufacture of a medicament for conditioning a subject prior to a transplant, as described herein.
  • an IL-2 agent described herein e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, in the manufacture of a medicament for conditioning a subject prior to a transplant, as described herein.
  • the disclosure provides use of an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, in the manufacture of a medicament for selectively increasing Tregs in a subject, as described herein, e.g., wherein the subject has received, is receiving, or will receive a transplant.
  • an IL-2 agent described herein e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein
  • the disclosure provides use of an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, in combination with a heart transplant, in the manufacture of a medicament for treating a heart disease or a symptom thereof in a subject, as described herein.
  • an IL-2 agent described herein e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein
  • FIG. 1A and IB are a series of flow cytometry plots showing that in the presence of TGF-P, an exemplary IL-2 mutein promoted murine inducible Treg (iTreg) development in vitro.
  • Treatment in the absence (FIG. 1A) or presence (FIG. IB) of 3 ng/ml TGFpi are indicated.
  • Treatment with beads only, IL-2/0.5 ug/ml, IL-2/5 ug/ml, IL-2m 0.5 ug/ml, or IL-2m 5 ug/ml are also indicated.
  • FIG. 2A and 2B are immunoblots showing that an exemplary IL-2 mutein promoted Stat5 tyrosine phosphorylation (Y 694) signaling from the IL-2R in Tregs.
  • FIG. 2A depicts Tyr-694 with p- Stat5b indicated by the arrow, and FIG. 2B depicts total. Lanes 1-4: beads only; 5-8: beads + IL-2 10 ug/ml; 9-12: beads + IL-2m 10 ug/ml. Fresh, 10, 30, and 60 min.
  • FIG. 3 is a series of flow cytometry plots showing expansion of circulating Tregs in vivo by injected an exemplary IL-2 mutein. 10 ug of IL-2 mutein were administered subcutaneously twice in a week. Analysis of blood pre-Tx is shown. X-axis indicates CD4 expression and y-axis indicates Foxp3 expression.
  • FIG. 4. is a graph showing the survival of a murine allograft model (BALB/c -> C57BL/6). Mice received RPM alone or two injections of IL-2 mutein pre-Tx, plus 0.5 mg/kg/d RPM for 14d (Alzet).
  • IL-2C exemplary IL-2 mutein
  • IL-2A mutein ⁇ 50% Tregs
  • IL-2C mutein Tregs ⁇ 30%
  • IL-2D mutein -20% Tregs -normal by d 10 post-Tx.
  • FIG. 6A is a graph showing the survival of a murine cardiac allograft model (BALB/c -> C57BL/6, 4-5/group) receiving RPM alone or IL-2C mutein (10 ug i.p. x2/wk IL-2C from Tx) with RPM (0.5 mg/kg/d RPM 14d (Alzet)) post-transplant. I1-2C mutein dosing was stopped at day 21 or continued to day 80 post-Tx. C3H cardiac allografts were received at 100 days. The results demonstrate that limited post-transplant IL-2C mutein prolonged murine cardiac allograft survival through the end of the study period.
  • FIG. 6B is a series of flow cytometry plots depicting CD4 expression (x-axis) and Foxp3 expression (y-axis), related to the murine cardiac allograft model and treatment shown in FIG. 6A.
  • the right-most flow cytometry plot corresponds to recipients with rejection.
  • the disclosure is based, at least in part, the discovery that a brief course of therapy with a selected IL-2 mutein can expand recipient Foxp3+ Treg cells and induce donor-specific allograft survival.
  • IL-2 muteins promote iTreg development in vitro and in vivo, and their efficacy is superior to that of native IL-2 in terms of promoting murine allograft survival.
  • pre-and post-treatment therapy prolonged allograft survival, especially when combined with a brief sub-therapeutic course of RPM.
  • the articles “a” and “an” refer to one or to more than one (e.g., to at least one) of the grammatical object of the article.
  • the term “or” is used herein to mean, and is used interchangeably with, the term “and/or”, unless context clearly indicates otherwise.
  • “About” and “approximately” shall generally mean an acceptable degree of error for the quantity measured given the nature or precision of the measurements. Exemplary degrees of error are within 20 percent (%), typically, within 10%, and more typically, within 5% of a given value or range of values. When “about” or “approximately” is present before a series of numbers or a range, it is understood that “about” or “approximately” can modify each of the numbers in the series or range.
  • compositions and methods disclosed herein encompass polypeptides and nucleic acids having the sequences specified, or sequences substantially identical or similar thereto, e.g, sequences at least 85%, 90%, 95% identical or higher to the sequence specified.
  • amino acid sequence in the context of an amino acid sequence, the term “substantially identical” is used herein to refer to a first amino acid that contains a sufficient or minimum number of amino acid residues that are i) identical to, or ii) conservative substitutions of aligned amino acid residues in a second amino acid sequence such that the first and second amino acid sequences can have a common structural domain and/or common functional activity.
  • amino acid sequences that contain a common structural domain having at least about 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity to a reference sequence, e.g, a sequence provided herein.
  • nucleotide sequence in the context of nucleotide sequence, the term “substantially identical” is used herein to refer to a first nucleic acid sequence that contains a sufficient or minimum number of nucleotides that are identical to aligned nucleotides in a second nucleic acid sequence such that the first and second nucleotide sequences encode a polypeptide having common functional activity, or encode a common structural polypeptide domain or a common functional polypeptide activity.
  • the term “functional variant” refers polypeptides that have a substantially identical amino acid sequence to the naturally -occurring sequence, or are encoded by a substantially identical nucleotide sequence, and are capable of having one or more activities of the naturally -occurring sequence.
  • the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes).
  • the length of a reference sequence aligned for comparison purposes is at least 30%, e.g., at least 40%, 50%, 60%, e.g, at least 70%, 80%, 90%, 100% of the length of the reference sequence.
  • the amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position.
  • the percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.
  • the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm.
  • the percent identity between two amino acid sequences is determined using the Needleman and Wunsch ((1970) J. Mol. Biol. 48:444-453) algorithm which has been incorporated into the GAP program in the GCG software package (available at www.gcg.com), using either a Blossum 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6.
  • the percent identity between two nucleotide sequences is determined using the GAP program in the GCG software package (available at www.gcg.com), using a NWSgapdna.CMP matrix and a gap weight of 40, 50, 60, 70, or 80 and a length weight of 1, 2, 3, 4, 5, or 6.
  • One suitable set of parameters are a Blossum 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5.
  • the percent identity between two amino acid or nucleotide sequences can be determined using the algorithm of E. Meyers and W. Miller ((1989) CABIOS, 4:11-17) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.
  • nucleic acid and protein sequences described herein can be used as a “query sequence” to perform a search against public databases to, for example, identify other family members or related sequences. Such searches can be performed using the NBLAST and XBLAST programs (version 2.0) of Altschul, et al. (1990) J. Mol. Biol. 215:403-10.
  • Gapped BLAST can be utilized as described in Altschul et al., (1997) Nucleic Acids Res. 25:3389-3402.
  • the default parameters of the respective programs e.g., XBLAST and NBLAST
  • the term “hybridizes under low stringency, medium stringency, high stringency, or very high stringency conditions” describes conditions for hybridization and washing. Guidance for performing hybridization reactions can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y.
  • hybridization conditions referred to herein are as follows: 1) low stringency hybridization conditions in 6X sodium chloride/sodium citrate (SSC) at about 45°C, followed by two washes in 0.2X SSC, 0.1% SDS at least at 50°C (the temperature of the washes can be increased to 55°C for low stringency conditions); 2) medium stringency hybridization conditions in 6X SSC at about 45°C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 60°C; 3) high stringency hybridization conditions in 6X SSC at about 45°C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 65°C; and preferably 4) very high stringency hybridization conditions are 0.5M sodium phosphate, 7% SDS at 65°C, followed by one or more washes at 0.2X SSC, 1%
  • amino acid is intended to embrace all molecules, whether natural or synthetic, which include both an amino functionality and an acid functionality and capable of being included in a polymer of naturally -occurring amino acids.
  • exemplary amino acids include naturally -occurring amino acids; analogs, derivatives and congeners thereof; amino acid analogs having variant side chains; and all stereoisomers of any of any of the foregoing.
  • amino acid includes both the D- or L- optical isomers and peptidomimetics.
  • a “conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain.
  • Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine).
  • polypeptide “peptide” and “protein” (if single chain) are used interchangeably herein to refer to polymers of amino acids of any length.
  • the polymer may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non-amino acids.
  • the terms also encompass an amino acid polymer that has been modified; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation, such as conjugation with a labeling component.
  • the polypeptide can be isolated from natural sources, can be a produced by recombinant techniques from a eukaryotic or prokaryotic host, or can be a product of synthetic procedures.
  • protein fragments, functional protein domains, and homologous proteins are also considered to be within the scope of this invention.
  • any protein fragment of a reference protein meaning a polypeptide sequence at least one amino acid residue shorter than a reference polypeptide sequence but otherwise identical
  • any protein that includes a stretch of about 20, about 30, about 40, about 50, or about 100 amino acids which are about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 98%, or about 100% identical to any of the sequences described herein can be utilized in accordance with the invention.
  • a protein sequence to be utilized in accordance with the disclosure includes 2, 3, 4, 5, 6, 7, 8, 9, 10, or more mutations as shown in any of the sequences provided or referenced herein.
  • nucleic acid refers to a polymeric form of nucleotides of any length, either deoxyribonucleotides or ribonucleotides, or analogs thereof.
  • the polynucleotide may be either single-stranded or double-stranded, and if single-stranded may be the coding strand or non-coding (antisense) strand.
  • a polynucleotide may comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs.
  • the sequence of nucleotides may be interrupted by non-nucleotide components.
  • a polynucleotide may be further modified after polymerization, such as by conjugation with a labeling component.
  • the nucleic acid may be a recombinant polynucleotide, or a polynucleotide of genomic, cDNA, semisynthetic, or synthetic origin which either does not occur in nature or is linked to another polynucleotide in a non-natural arrangement.
  • isolated refers to material that is removed from its original or native environment (e.g., the natural environment if it is naturally occurring).
  • a naturally -occurring polynucleotide or polypeptide present in a living animal is not isolated, but the same polynucleotide or polypeptide, separated by human intervention from some or all of the coexisting materials in the natural system, is isolated.
  • Such polynucleotides could be part of a vector and/or such polynucleotides or polypeptides could be part of a composition, and still be isolated in that such vector or composition is not part of the environment in which it is found in nature.
  • a disorder e.g, a myeloma
  • a subject e.g., a human
  • a disorder e.g., a myeloma
  • a symptom of a disorder e.g, a myeloma
  • a myeloma when a myeloma is treated, a bone marrow biopsy will show fewer clonal plasma cells, after effective treatment for myeloma.
  • a diagnostic assay will detect fewer clonal plasma cells in a biological sample of a subject after administration of an antibody molecule described herein for the effective treatment of a myeloma.
  • Other assays, urine tests, or blood tests can also be used to monitor treatment in a patient, or to detect the presence, e.g., decreased presence (or absence), of a symptom of a myeloma, after treatment of a myeloma in the subject.
  • the level of P2 microglobulin (P2M) in serum or urine will be decreased, after effective treatment for myeloma.
  • Treatment can, e.g, partially or completely, alleviate, ameliorate, relieve, inhibit, or reduce the severity of, and/or reduce incidence, and optionally, delay onset of, one or more manifestations of the effects or symptoms, features, and/or causes of a disorder, e.g., a myeloma.
  • treatment is of a subject who does not exhibit certain signs of a disorder, e.g., a myeloma, and/or of a subject who exhibits only early signs of a disorder, e.g., nephropathy.
  • treatment is of a subject who exhibits one or more established signs of a disorder, e.g, a myeloma.
  • treatment is of a subject diagnosed as suffering from a disorder, e.g, a myeloma.
  • a disorder e.g, a myeloma
  • a subject e.g., a human
  • the disorder e.g, a myeloma, if the subject receives the antibody molecule.
  • IL-2 agents including, but not limited to, IL-2 variants, IL-2 fusion proteins, IL-2 complexes, and IL-2 conjugates.
  • the IL-2 agents described herein can have one or more structural and/or functional properties described herein.
  • the IL-2 agent comprises an IL-2 variant comprising one or more amino acid alterations (e.g., substitutions) described herein.
  • the IL-2 agent comprises an IL-2 variant comprising one or more amino acid alterations (e.g., substitutions) described in Table 9.
  • the IL-2 agent comprises an IL-2 variant comprising an amino acid sequence described in Table 9, or a portion thereof.
  • the IL-2 agent is encoded by a nucleic acid comprising a nucleotide sequence described herein, e.g., in Table 10.
  • the one or more amino acid alterations e.g., substitutions
  • the IL-2 agent can modulate (e.g., increase) Treg proliferation, survival, activation and/or function.
  • the modulation is selective or specific for the Tregs.
  • the IL-2 agent is capable of modulating the activity in Tregs but has limited or lacks the ability to promote the activity in non-regulatory T cells.
  • the IL-2 agent comprises a polypeptide (sometime referred to herein as “IL-2 polypeptide agent”). IL-2 Variants
  • the IL-2 agent comprises an IL-2 variant, e.g., an IL-2 variant described herein.
  • the IL-2 variant comprises an IL-2 polypeptide (e.g., a human IL-2 polypeptide) described herein, or a functional fragment thereof.
  • the IL-2 variant comprises one or more amino acid alterations (e.g., substitutions) described in Table 9.
  • the IL-2 variant comprises, or consists of, an amino acid sequence described in Table 9, or a functional fragment thereof.
  • the IL-2 variant is encoded by a nucleic acid comprising a nucleotide sequence described herein, e.g, in Table 10.
  • the IL-2 variants described herein which have reduced human CD25 and/or reduced human CD122/CD132 binding affinity relative to a wild-type human IL-2 or a reference IL-2 variant, can have improved potency and/or selectivity for binding to and activating regulatory T cells (Tregs) than wild type IL-2 or other IL-2 variants.
  • the IL-2 variants described herein can be identified, e.g, by screening a library of mutated IL-2 polypeptides to identify IL-2 variants having a binding affinity for human CD25 and/or human CD122/CD132 in a desired range.
  • the IL-2 variant has one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more) properties described herein, e.g, different and/or improved properties, relative to a wild-type IL-2 or a reference IL-2 variant.
  • the IL-2 variant comprises one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) amino acid alterations (e.g., substitutions) that provide different and/or improved properties, relative to a wild-type IL-2 or a reference IL-2 variant.
  • the IL-2 variant has one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or all) of the following different and/or improved properties (e.g., as determined by an assay described herein), relative to a wild-type IL-2 or a reference IL-2 variant: i) altered (e.g., enhanced or increased) expression in vitro and/or in vivo', ii) altered (e.g., reduced or decreased) aggregation in vitro and/or in vivo-, iii) altered (e.g., enhanced or increased) stability in vitro and/or in vivo-, iv) altered (e.g., enhanced or increased) half-life in vitro and/or in vivo; v) altered (e.g., reduced or decreased) turnover and/or clearance in vivo; vi) altered (e.g., reduced or decreased) susceptibility to proteolysis in vitro and/or in vivo; vii) altered (e.
  • the IL-2 variant has altered (e.g., enhanced or increased) expression in vitro and/or in vivo, relative to a wild-type IL-2 or a reference IL-2 variant.
  • the IL-2 variant has enhanced or increased expression (e.g., in a bacterial or mammalian cell) relative to a wild-type IL-2.
  • the IL-2 variant has enhanced or increased expression (e.g., in bacterial or mammalian cell) relative to a reference IL-2 variant.
  • the expression of the IL-2 variant is increased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more.
  • the expression of the IL-2 variant is increased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or about 10-fold, or more.
  • the IL-2 variant expresses at a higher or increased level in vitro and/or in vivo, e.g., increased by about 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100% or more e.g.
  • the IL-2 variant expresses at a higher or increased level, e.g, increased by about 0.5-fold, about 1-fold, about 1.5-fold, about 2-fold, about 2.5-fold, about 3-fold, about 3.5-fold, about 4-fold, about 4.5-fold, about 5-fold, about 5.5-fold, about 6-fold, about 6.5-fold, about 7-fold, about 7.5-fold, about 8-fold, about 8.5 -fold, about 9-fold, about 9.5 -fold, about 10-fold or more e.g, relative to an IL-2 agent comprising a wild-type IL-2 or an IL-2 agent comprising a reference IL-2 variant e.g., as determined by an assay of protein concentration.
  • the IL-2 variant has altered (e.g., reduced or decreased) aggregation in vitro and/or in vivo, relative to a wild-type IL-2 or a reference IL-2 variant. In an embodiment, the IL-2 variant has reduced or decreased aggregation relative to a wild type IL-2. In an embodiment, the IL-2 variant has reduced or decreased aggregation relative to a reference IL-2 variant. In an embodiment, the aggregation of the IL-2 variant is decreased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more.
  • the aggregation of the IL-2 variant is decreased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or about 10-fold, or more.
  • an IL-2 agent comprising an IL-2 variant described herein aggregates at lower or decreased level in vitro and/or in vivo, e.g., decreased by about 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100% or more e.g., relative to an IL-2 agent comprising a wild-type IL-2 or an IL-2 agent comprising a reference IL-2 variant e.g., as determined by melting temperature analysis (e.g., using fluorimetry), dynamic
  • an IL-2 agent comprising an IL-2 variant described herein aggregates at lower or decreased level, e.g., decreased by about 0.5-fold, about 1- fold, about 1.5-fold, about 2-fold, about 2.5-fold, about 3-fold, about 3.5-fold, about 4-fold, about 4.5- fold, about 5-fold, about 5.5-fold, about 6-fold, about 6.5-fold, about 7-fold, about 7.5-fold, about 8- fold, about 8.5 -fold, about 9-fold, about 9.5 -fold, about 10-fold or more e.g, relative to an IL-2 agent comprising a wild-type IL-2 or an IL-2 agent comprising a reference IL-2 variant, e.g., as determined by melting temperature analysis (e.g., using fluorimetry), dynamic light scattering, and/or sizeexclusion chromatography.
  • melting temperature analysis e.g., using fluorimetry
  • dynamic light scattering e.g., dynamic light scattering
  • sizeexclusion chromatography e.g
  • the IL-2 variant has altered (e.g., enhanced or increased) stability in vitro and/or in vivo, relative to a wild-type IL-2 or a reference IL-2 variant.
  • the IL-2 variant has enhanced or increased stability relative to a wild-type IL-2.
  • the IL-2 variant has enhanced or increased stability relative to a reference IL-2 variant.
  • the stability of the IL-2 variant is increased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more.
  • the stability of the IL-2 variant is increased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or about 10-fold, or more.
  • an IL-2 agent comprising an IL-variant described herein has enhanced or increased stability in vitro and/or in vivo, e.g., increased by about 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100% or more, or e.g., increased by about 0.5- fold, about 1-fold, about 1.5-fold, about 2-fold, about 2.5-fold, about 3-fold, about 3.5-fold, about 4- fold, about 4.5-fold, about 5-fold, about 5.5-fold, about 6-
  • the IL-2 variant has altered (e.g., enhanced or increased) half-life in vitro and/or in vivo, relative to a wild-type IL-2 or a reference IL-2 variant.
  • the IL-2 variant has enhanced or increased half-life relative to a wild-type IL-2.
  • the IL-2 variant has enhanced or increased half-life relative to a reference IL-2 variant.
  • the half-life of the IL-2 variant is increased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more.
  • the half-life of the IL-2 variant is increased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or about 10-fold, or more.
  • an IL-2 agent comprising an IL-2 variant described herein has enhanced or increased half-life in vitro and/or in vivo, e.g., increased by about 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100% or more, or e.g., greater than about 0.5- fold, about 1-fold, about 1.5-fold, about 2-fold, about 2.5-fold, about 3-fold, about 3.5-fold, about 4- fold, about 4.5-fold, about 5-fold, about 5.5-fold
  • the IL-2 variant has altered (e.g., reduced or decreased) turnover in vitro and/or in vivo, relative to a wild-type IL-2 or a reference IL-2 variant. In an embodiment, the IL-2 variant has reduced or decreased turnover relative to a wild-type IL-2. In an embodiment, the IL-2 variant has reduced or decreased turnover relative to a reference IL-2 variant. In an embodiment, the turnover of the IL-2 variant is decreased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more.
  • the turnover of the IL-2 variant is decreased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, about 10-fold, or more.
  • an IL-2 agent comprising an IL-2 variant described herein has a lower, reduced or decreased rate or level of turnover and/or clearance in vivo, e.g, decreased by about 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100% or more, or e.g., decreased by about 0.5-fold, about 1-fold, about 1.5-fold, about 2-fold, about 2.5-fold, about 3-fold, about 3.5-fold, about 4-fold, about 4.5-fold, about 5-fold, about 5.5-fold
  • the IL-2 has altered (e.g., reduced or decreased) susceptibility to proteolysis in vitro and/or in vivo, relative to a wild-type IL-2 or a reference IL-2 variant.
  • the IL-2 variant has reduced or decreased susceptibility to proteolysis relative to IL-2 (e.g., wild type human IL-2).
  • the IL-2 variant has reduced or decreased susceptibility to proteolysis relative to a reference IL-2 variant.
  • the susceptibility to proteolysis of the IL-2 variant is decreased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more.
  • the susceptibility to proteolysis of the IL-2 variant is decreased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8- fold, 9-fold, or about 10-fold, or more.
  • the IL-2 variant has altered (e.g., enhanced or increased) resistance to proteolysis in vitro and/or in vivo, relative to a wild-type IL-2 or a reference IL-2 variant.
  • the IL-2 variant has enhanced or increased resistance to proteolysis relative to a wildtype IL-2.
  • the IL-2 variant has enhanced or increased resistance to proteolysis relative to a reference IL-2 variant.
  • the resistance to proteolysis of the IL-2 variant is increased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more.
  • the resistance to proteolysis of the IL-2 variant is increased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or about 10-fold, or more.
  • the IL-2 variant has altered (e.g., reduced or decreased) binding capacity and/or binding affinity for human CD25 in vitro and/or in vivo, relative to a wild-type IL-2 or a reference IL-2 variant. In an embodiment, the IL-2 variant has reduced or decreased binding capacity and/or binding affinity for human CD25 relative to a wild-type human IL-2). In an embodiment, the IL-2 variant has reduced or decreased binding capacity and/or binding affinity for human CD25 relative to a reference IL-2 variant.
  • the binding capacity and/or binding affinity of the IL-2 variant for human CD25 is decreased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more. In an embodiment, the binding capacity and/or binding affinity of the IL-2 variant for human CD25 is decreased by about 0.5-fold, 1-fold, 2-fold, 3- fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or about 10-fold, or more.
  • an IL- 2 agent comprising an IL-2 variant described herein has reduced or decreased binding affinity for CD25 (e.g., human CD25), e.g, decreased by about 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100% or more, or e.g, decreased by about 0.5-fold, about 1-fold, about 1.5-fold, about 2-fold, about 2.5-fold, about 3-fold, about 3.5-fold, about 4-fold, about 4.5-fold, about 5-fold, about 5.5-fold, about 6-fold, about 6.5-fold, about 7-fold, about 7.5-fold, about 8-fold, about 8.5-fold, about 9-fold, about 9.5-fold, about 10-fold or more e.g., relative to an IL-2
  • the IL-2 variant binds to CD25 (e.g., human CD25) with low affinity, e.g., with a dissociation constant (K D ) of about 5-500 pM, e.g, about 5, about 10, about 15, about 20, about 25, about 30, about 35, about 40, about 45, about 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 105, about 110, about 115, about 120, about 125, about 130, about 135, about 140, about 145, about 150, about 200, about 250, about 300, about 350, about 400, about 450, or about 500 pM, or e.g., about 10 to about 400 pM, about 20 to about 300 pM, about 50 to about 200 pM, about 100 to about 150 pM, about 5 to about 10 pM, e.g., about 10 to about 20 pM, about 20 to about 30 pM, or about 30 to about
  • the IL-2 variant binds to CD25 (e.g., human CD25) with low affinity, e.g., with a dissociation constant (K D ) of about 0.1-10 nM, e.g., about 0.1, about 0.2, about 0.3, about 0.4, about 0.5, about 0.6, about 0.7, about 0.8, about 0.9, about 1, about 1.5, about 2, about 2.5, about 3, about 3.5, about 4, about 4.5, about 5, about 6, about 7, about 8, about 9, or about 10 nM, or e.g., about 0.2 to about 5 nM, about 0.5 to about 2 nM, about 1 to 1.5 nM, about 0.1 to about 0.2 nM, e.g., about 0.2 to about 0.3 nM, about 0.3 to about 0.4 nM, or about 0.4 to about 0.5 nM, e.g., about 0.5 to about 0.6 nM, about 0.6 to about 0.7 nM, about 0.7 to
  • nM e.g., as determined by surface plasmon resonance (e.g. Biacore) and/or bio-layer interferometry (e.g., Octet binding).
  • surface plasmon resonance e.g. Biacore
  • bio-layer interferometry e.g., Octet binding
  • the IL-2 variant has altered (e.g., reduced or decreased) binding capacity and/or binding affinity for human CD 132 in vitro and/or in vivo, relative to a wild-type IL-2 or a reference IL-2 variant. In an embodiment, the IL-2 variant has reduced or decreased binding capacity and/or binding affinity for human CD 132 relative to a wild-type IL-2. In an embodiment, the IL-2 variant has reduced or decreased binding capacity and/or binding affinity for human CD 132 relative to a reference IL-2 variant.
  • the binding capacity and/or binding affinity of the IL-2 variant for human CD132 is decreased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more. In an embodiment, the binding capacity and/or binding affinity of the IL-2 variant for human CD132 is decreased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4- fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or about 10-fold, or more.
  • the IL-2 variant has altered (e.g., reduced or decreased) binding capacity and/or binding affinity for the human dimeric IL-2 receptor comprising human CD 122 and human CD 132 in vitro and/or in vivo, relative to a wild-type IL-2 or a reference IL-2 variant.
  • the IL-2 variant has reduced or decreased binding capacity and/or binding affinity for the human dimeric IL-2 receptor comprising human CD122 and human CD132 relative to a wild-type IL-2.
  • the IL-2 variant has reduced or decreased binding capacity and/or binding affinity for the human dimeric IL-2 receptor comprising human CD 122 and human CD 132 relative to a reference IL-2 variant.
  • the binding capacity and/or binding affinity of the IL-2 variant for the human dimeric IL-2 receptor comprising human CD122 and human CD132 is decreased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more. In an embodiment, the binding capacity and/or binding affinity of the IL-2 variant for the human dimeric IL-2 receptor comprising human CD122 and human CD132 is decreased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or about 10-fold, or more.
  • the IL-2 variant has reduced or decreased binding affinity for CD122/CD132 heterodimer (e.g., human CD122/CD132 heterodimer), e.g., decreased by about 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100% or more, or e.g., decreased by about 0.5-fold, about 1-fold, about 1.5-fold, about 2-fold, about 2.5-fold, about 3-fold, about 3.5-fold, about 4-fold, about 4.5-fold, about 5-fold, about 5.5-fold, about 6-fold, about 6.5-fold, about 7-fold, about 7.5-fold, about 8-fold, about 8.5-fold, about 9-fold, about 9.5-fold, about 10-fold or more e.g, relative to
  • the IL-2 variant binds to CD 122/CD 132 heterodimer (e.g., human CD122/CD132 heterodimer) with low affinity, e.g., with a dissociation constant (KD) of about 0.2-20 nM, e.g, about 0.2, about 0.3, about 0.4, about 0.5, about 0.6, about 0.7, about 0.8, about 0.9, about 1, about 1.1, about 1.2, about 1.3, about 1.4.
  • KD dissociation constant
  • nM about 1.5, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, or about 20 nM, or e.g, about 0.5 to about 15 nM, about 1 to about 10 nM, about 2 to about 5 nM, about 0.2 to about 0.3 nM, about 0.3 to about 0.4 nM, about 0.4 to about 0.5 nM, about 0.5 to about 0.6 nM, about 0.6 to about 0.7 nM, about 0.7 to about 0.8 nM, about 0.8 to about 0.9 nM, about 0.9 to about 1 nM, about 1 to about 1.1 nM, about 1.1 to about 1.2 nM, about 1.2 to about 1.3 nM, about 1.3 to about 1.4 nM, about 1.4 to about 1.5 nM, about 1.5 to about 2 nM, about 2 to about 3 nM, about 3 to about 4 nM, about 4 to about 5
  • the IL-2 variant binds to CD 122/CD 132 heterodimer (e.g., human CD122/CD132 heterodimer) with low affinity, e.g., with a dissociation constant (KD) of about 0.2- 300 nM , e.g., about 0.2 nM, about 0.5 nM, about 1 nM, about 2 nM, about 5 nM, about 10 nM, about 15 nM, about 20 nM, about 25 nM, about 30 nM, about 40 nM, about 50 nM, about 60 nM, about 70 nM, about 80 nM, about 90 nM, about 100 nM, about 110 nM, about 120 nM, about 130 nM
  • KD dissociation constant
  • the IL-2 variant has altered (e.g., enhanced, increased, and/or selective) binding to Tregs in vitro and/or in vivo, relative to a wild-type IL-2 or a reference IL-2 variant.
  • the IL-2 variant has enhanced or increased binding to Tregs relative to a wild-type IL-2.
  • the IL-2 variant has selective binding to Tregs relative to IL-2 (e.g., wild type human IL-2).
  • the IL-2 variant has enhanced or increased binding to Tregs relative to a reference IL-2 variant.
  • the IL-2 variant has selective binding to Tregs relative to a reference IL-2 variant.
  • the binding to Tregs is increased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more. In an embodiment, the binding to Tregs is increased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7- fold, 8-fold, 9-fold, or about 10-fold, or more.
  • the IL-2 variant has altered (e.g., enhanced, increased, and/or selective) activation of the IL-2 signaling pathway in Tregs in vitro and/or in vivo, relative to a wild-type IL-2 or a reference IL-2 variant.
  • the IL-2 variant has enhanced or increased activation of the IL-2 signaling pathway in Tregs relative to a wild-type IL-2. In an embodiment, the IL-2 variant has selective activation of the IL-2 signaling pathway in Tregs relative to a wild-type IL-2. In an embodiment, the IL-2 variant has enhanced or increased activation of the IL-2 signaling pathway in Tregs relative to a reference IL-2 variant. In an embodiment, the IL-2 variant has selective activation of the IL-2 signaling pathway in Tregs relative to a reference IL-2 variant.
  • the activation of the IL-2 signaling pathway in Tregs is increased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more. In an embodiment, the activation of the IL-2 signaling pathway in Tregs is increased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5- fold, 6-fold, 7-fold, 8-fold, 9-fold, or about 10-fold, or more.
  • the IL-2 variant selectively activates IL-2 signaling in T regulatory cells in vitro and/or in vivo, e.g., having an T helper EC50/Treg EC50 ratio greater than about 1, about 2, about 3, about 4, about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1000, 1500, 2000, 2500, or about 3000 or more relative to an IL-2 agent comprising a wild-type IL-2 or an IL-2 agent comprising a reference IL-2 variant e.g., as determined flow cytometry.
  • the IL-2 variant selectively activates IL-2 signaling in T regulatory cells in vitro and/or in vivo, e.g., having an NK cell EC50/Treg EC50 ratio greater than e.g, about 1, about 2, about 3, about 4, about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1000, 1500, 2000, 2500, or about 3000 or more, or e.g., greater than 1 and about 1 to 2, about 2 to 3, about 3 to 4, about 4 to 5, greater than 1 and about 1 to 10, greater than 1 and about 1 to 20, greater than 1 and about 1 to 30, greater than 1 and about 1 to 40, greater than 1 and about 1 to 50, about 2 to 10, about 2 to 20, about 2 to 30, about 2 to 40, 2 to
  • the IL-2 variant has altered (e.g., enhanced, increased, and/or selective) ability to induce or promote Treg expansion, activity, survival, and/or proliferation in vitro and/or in vivo, relative to a wild-type IL-2 or a reference IL-2 variant.
  • the IL-2 variant has enhanced or increased ability to induce or promote Treg expansion, activity, survival, and/or proliferation relative to a wild-type IL-2.
  • the IL-2 variant has selective ability to induce or promote Treg expansion, activity, survival, and/or proliferation relative to a wild-type IL-2.
  • the IL-2 variant has enhanced or increased ability to induce or promote Treg expansion, activity, survival, and/or proliferation relative to a reference IL-2 variant. In an embodiment, the IL-2 variant has selective ability to induce or promote Treg expansion, activity, survival, and/or proliferation relative to a reference IL-2 variant. In an embodiment, the ability to induce or promote Treg expansion, activity, survival, and/or proliferation is increased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more.
  • the ability to induce or promote Treg expansion, activity, survival, and/or proliferation is increased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or about 10-fold, or more.
  • the IL-2 variant has enhanced or increased potency and/or ability to induce or promote T regulatory cell activity, e.g., having an EC50 for Tregs that is lower by about 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100% or more, or e.g., decreased by about 0.5- fold, about 1-fold, about 1.5-fold, about 2-fold, about 2.5-fold, about 3-fold, about 3.5-fold, about 4- fold, about 4.5-fold, about 5-fold, about 5.5-fold, about 6-fold, about 6.5-fold, about 7-fold, about 7.5- fold, about 8-fold, about 8.5-fold, about 9-fold, about 9.5-fold, about 10-fold or more e.g, relative to an IL
  • the IL-2 variant has reduced or decreased potency and/or ability to induce or promote T regulatory cell activity, e.g, having an EC50 for Tregs that is higher by about 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% or more, or e.g, decreased by about 0.5-fold, about 1-fold, about 1.5-fold, about 2-fold, about 2.5-fold, about 3-fold, about 3.5-fold, about 4-fold, about 4.5-fold, about 5-fold, about 5.5-fold, about 6-fold, about 6.5-fold, about 7-fold, about 7.5-fold, about 8-fold, about 8.5-fold, about 9-fold, about 9.5-fold, about 10-fold, about 50-fold, about 100-fold, about 200
  • the T helper cell described herein is a CD45+CD3+CD4+Foxp3- cell, e.g., determined by flow cytometry.
  • the Treg described herein is CD45+CD3+CD4+Foxp3+ cell, e.g., determined by flow cytometry.
  • the NK cell described herein is a CD45+CD3- cell that is CD56+ and/or CD16+, e.g., determined by flow cytometry.
  • the NK cell described herein is a CD45+CD3-CD56+ cell, e.g., determined by flow cytometry.
  • the IL-2 variant has one or more of the same, or substantially the same, structural and/or functional properties, as a wild-type IL-2 or a reference IL-2 variant.
  • the reference IL-2 variant comprises an amino acid sequence that has about 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity to an IL-2 variant described herein.
  • the reference IL-2 variant comprises the amino acid sequence of SEQ ID NO: 1 (IL-2 C125S).
  • the IL-2 variant comprises an amino acid sequence that is at least 80%, 85%, 90%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO: 1 and comprises one or more (2, 3, 4, 5, 6, 7, 8, 9, 10, or more) amino acid alterations (e.g., substitutions) described herein.
  • IL-2 variant position numbering begins at the first amino acid following the signal peptide of the exemplary wild type (WT) human IL-2 polypeptide: MYRMQLLSCIALSLALVTNS/A1/P2/T3/S4/S5/S6/T7/K8/K9/T 10/Q 11/L 12/Q 13/L 14/E 15/H 16/L 1 7/L18/L19/D20/L21/Q22/M23/I24/L25/N26/G27/I28/N29/N30/Y31/K32/N33/P34/K35/L36/T37/R38 /M39/L40/T41/F42/K43/F44/Y45/M46/P47/K48/K49/A50/T51/E52/L53/K54/H55/L56/Q57/C58/L59 /E60/E61/E62/L63/K64/P65/L66/E67
  • the IL-2 agent comprises amino acid alteration(s) (e.g., substitution(s)) at position(s) corresponding to human IL-2 (e.g., comprising the amino acid sequence of SEQ ID NO: 1031).
  • the IL-2 variant comprises the amino acid sequence of A1/P2/X3/S4/S5/S6/T7/K8/K9/T 10/Q 11/L 12/Q 13/L 14/E 15/X 16/L 17/L 18/L 19/D20/L21/Q22/M23/I2 4/L25/N26/G27/X28/N29/N30/Y31/K32/N33/P34/X35/L36/T37/X38/M39/L40/T41/X42/K43/F44/Y 45/M46/P47/K48/K49/A50/T51/E52/L53/K54/H55/L56/Q57/C58/L59/E60/E61/E62/L63/K64/P65/L 66/E67/X68/X69/L70/N71/L72/A73/X74/S75/K76/N77/F78/H79/L80/R
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or all) of positions, as described herein.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or all) of positions chosen from T3, H16, 128, K35, R38, F42, E68, V69, Q74, D84, S87, N88, 192, C125, or Q126.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position T3. In an embodiment, the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position Hl 6. In an embodiment, the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position 128. In an embodiment, the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position K35. In an embodiment, the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position R38. In an embodiment, the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position F42.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position E68. In an embodiment, the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position V69. In an embodiment, the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position Q74. In an embodiment, the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position D84. In an embodiment, the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position S87. In an embodiment, the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position N88.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position 192. In an embodiment, the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position Cl 25. In an embodiment, the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position Q126.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position V69, Q74, or a combination thereof. In an embodiment, the IL-2 variant comprises an amino acid alteration (e.g., substitution) at positions V69 and Q74. In an embodiment, the IL-2 variant comprises the amino acid substitution V69A. In an embodiment, the IL-2 variant comprises the amino acid substitution Q74P.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position H16, 192, D84, or a combination thereof. In an embodiment, the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position Hl 6, optionally wherein the amino acid substitution is H16N, H16L, or H16D. In an embodiment, the IL-2 variant comprises the amino acid substitution H16N. In an embodiment, the IL-2 variant comprises the amino acid substitution H16L. In an embodiment, the IL-2 variant comprises the amino acid substitution H16D. In an embodiment, the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position at 192, optionally wherein the amino acid substitution is I92S. In an embodiment, the IL-2 variant comprises the amino acid substitution I92S.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position D84, optionally wherein the amino acid substitution is D84V. In an embodiment, the IL-2 variant comprises the amino acid substitution is D84V.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position K35, R38, F42, E68, or a combination thereof. In an embodiment, the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position K35, optionally wherein the amino acid substitution is K35E. In an embodiment, IL-2 variant comprises the amino acid substitution K35E.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position R38, optionally wherein the amino acid substitution is R38E, R38N or R38Q. In an embodiment, the IL-2 variant comprises the amino acid substitution R38N. In an embodiment, the IL-2 variant comprises the amino acid substitution R38Q.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position F42, optionally wherein the amino acid substitution is F42K or F42Q. In an embodiment, the IL-2 variant comprises the amino acid substitution F42K. In an embodiment, the IL-2 variant comprises the amino acid substitution F42Q.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution): (i) at (a) positions V69 and Q74, (b) position K35, or (c) positions V69, Q74, and K35; and (ii) at one, two, or all of positions Hl 6, 192, or D84.
  • the IL-2 variant further comprises an amino acid alteration (e.g., substitution) at one, two, or all of positions R38, F42, or E68.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution): (i) at (a) positions V69 and Q74, (b) position K35, or (c) positions V69, Q74, and K35; and (ii) at (a) one, two, or all of positions H16, 192, or D84; or (b) one, two, or all of positions R38, F42, or E68.
  • amino acid alteration e.g., substitution
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution): (i) at (a) positions V69 and Q74, (b) position K35, or (c) positions V69, Q74, and K35; and (ii) at (a) one, two, or all of positions H16, 192, or D84; and (b) one, two, or all of positions R38, F42, or E68.
  • amino acid alteration e.g., substitution
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position V69, Q74, and H16, optionally wherein the amino acid substitution is V69A, Q74P, and H16N or H16L, respectively.
  • the IL-2 variant comprises the amino acid substitutions V69A, Q74P, and H16N or H16L.
  • the IL-2 variant comprises the amino acid substitutions V69A, Q74P, and H16N.
  • the IL-2 variant comprises the amino acid substitutions V69A, Q74P, and H16L.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position V69, Q74, and 192, optionally wherein the amino acid substitution is V69A, Q74P, and I92S, respectively. In an embodiment, the IL-2 variant comprises the amino acid substitutions V69A, Q74P, and I92S.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position V69, Q74, and D84, optionally wherein the amino acid substitution is V69A, Q74P, and D84V, respectively.
  • the IL-2 variant comprises the amino acid substitutions V69A, Q74P, and D84V.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position V69, Q74, and R38, optionally wherein the amino acid substitution is V69A, Q74P, and R38Q, respectively.
  • the IL-2 variant comprises the amino acid substitutions V69A, Q74P, and R38Q.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position V69, Q74, and F42, optionally wherein the amino acid substitution is V69A, Q74P, and F42Q, respectively.
  • the IL-2 variant comprises the amino acid substitutions V69A, Q74P, and F42Q.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position V69, Q74, and R38, optionally wherein the amino acid substitution is V69A, Q74P, and R38N, respectively. In an embodiment, the IL-2 variant comprises the amino acid substitutions V69A, Q74P, and R38N.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position V69, Q74, and R38, optionally wherein the amino acid substitution is V69A, Q74P, and R38E, respectively.
  • the IL-2 variant comprises the amino acid substitution V69A, Q74P, and R38E.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position V69, Q74, K35, and H16, optionally wherein the amino acid substitution is V69A, Q74P, K35E, and H16N, respectively.
  • the IL-2 variant comprises the amino acid substitutions V69A, Q74P, K35E, and H16N.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position V69, Q74, K35, H16, and R38, optionally wherein the amino acid substitution is V69A, Q74P, K35E, H16N, and R38N, respectively.
  • the IL-2 variant comprises the amino acid substitutions V69A, Q74P, K35E, H16N, and R38N.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position V69, Q74, H16, and R38, optionally wherein the amino acid substitution is V69A, Q74P, H16N, and R38N or R38Q, respectively.
  • the IL-2 variant comprises the amino acid substitutions V69A, Q74P, H16N, and R38N or R38Q.
  • the IL-2 variant comprises the amino acid substitutions V69A, Q74P, H16N, and R38N.
  • the IL-2 variant comprises the amino acid substitutions V69A, Q74P, H16N, and R38Q.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position 128, E68, S87, N88, Q126, or a combination thereof.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position 128, optionally wherein the amino acid substitution is I28T or I28F. In an embodiment, the IL-2 variant comprises the amino acid substitution I28T. In an embodiment, the IL-2 variant comprises the amino acid substitution I28F.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position E68, optionally wherein the amino acid substitution is E68Q or E68N. In an embodiment, the IL-2 variant comprises the amino acid substitution E68Q. In an embodiment, the IL-2 variant comprises the amino acid substitution E68N.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position S87, optionally wherein the amino acid substitution is S87R. In an embodiment, the IL-2 variant comprises the amino acid substitution S87R.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position N88, optionally wherein the amino acid substitution is N88S, N88L, or N88D. In an embodiment, the IL-2 variant comprises the amino acid substitution N88S, N88L, or N88D. In an embodiment, the IL-2 variant comprises the amino acid substitution N88S. In an embodiment, the IL- 2 variant comprises the amino acid substitution N88L. In an embodiment, the IL-2 variant comprises the amino acid substitution N88D.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position Q126, optionally wherein the amino acid substitution is Q126T, Q126K, or Q126R.
  • the IL-2 variant comprises the amino acid substitution Q126T, Q126K, or Q126R.
  • the IL-2 variant comprises the amino acid substitution Q126T, Q126K, or Q126R.
  • the IL-2 variant comprises the amino acid substitution Q126T.
  • the IL-2 variant comprises the amino acid substitution Q126K.
  • the IL-2 variant comprises the amino acid substitution Q126R.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position C125, optionally wherein the amino acid substitution is C125S. In an embodiment, the IL-2 variant comprises the amino acid substitution C125S.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position T3, optionally wherein the amino acid substitution is T3A. In an embodiment, the IL-2 variant comprises the amino acid substitution T3A.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position V69, Q74, and C125, optionally wherein the amino acid substitution is V69A, Q74P, and C125S, respectively. In an embodiment, the IL-2 variant comprises the amino acid substitutions V69A, Q74P, and C125S. In an embodiment, the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position T3, H16, 192, or a combination thereof, optionally wherein the amino acid substitution is T3A, H16N, and I92S, respectively.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position H16, V69, Q74, and C125, optionally wherein the amino acid substitution is H16N, V69A, Q74P, and C125S, respectively.
  • the IL-2 variant comprises the amino acid substitutions H16N, V69A, Q74P, and C125S.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position H16, V69, Q74, and C125, optionally wherein the amino acid substitution is H16L, V69A, Q74P, and C125S, respectively.
  • the IL-2 variant comprises the amino acid substitutions H16L, V69A, Q74P, and C125S.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position H16, V69, Q74, 192, and C125, optionally wherein the amino acid substitution is H16L, V69A, Q74P, I92S, and C125S, respectively.
  • the IL-2 variant comprises the amino acid substitutions H16L, V69A, Q74P, I92S, and C125S.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position T3, V69, Q74, and C125, optionally wherein the amino acid substitution is T3A, V69A, Q74P, and C125S, respectively.
  • the IL-2 variant comprises the amino acid substitutions T3A, V69A, Q74P, and C125S.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position T3, H16, V69, Q74, and C125, optionally wherein the amino acid substitution is T3A, H16N or H16L, V69A, Q74P, and C125S, respectively.
  • the IL-2 variant comprises the amino acid substitutions T3A, H16N, V69A, Q74P, and C125S.
  • the IL-2 variant comprises the amino acid substitutions T3A, H16L, V69A, Q74P, and C125S.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position T3, V69, Q74, 192, and C125, optionally wherein the amino acid substitution is T3A, V69A, Q74P, I92S, and C125S, respectively.
  • the IL-2 variant comprises the amino acid substitutions T3A, V69A, Q74P, I92S, and C125S.
  • the IL-2 variant comprises the amino acid substitutions T3A, V69A, Q74P, I92S, and C125S.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position H16, K35, V69 and Q74, optionally wherein the amino acid substitution is H16L, K35E, V69A, and Q74P, respectively.
  • the IL-2 variant comprises the amino acid substitutions H16L, K35E, V69A, and Q74P.
  • the IL-2 variant comprises an amino acid alteration (e.g., substitution) at position H16, R38, V69A, and Q74P, optionally wherein the amino acid substitution is H16L, R38Q, V69A, and Q74P, respectively.
  • the IL-2 variant comprises the amino acid substitutions H16L, R38Q, V69A, and Q74P.
  • the IL-2 variant comprises amino acid substitutions H16L, V69A, Q74P, and C125S.
  • the IL-2 variant comprises amino acid substitutions H16N, V69A, Q74P, and C125S.
  • an IL-2 variant comprising the aforesaid mutations also has reduced binding affinity for CD122 and/or CD132, which increases the potency and selectivity of the IL-2 variant for regulatory T cells (Treg) compared to other T cell types.
  • an IL-2 variant comprising the aforesaid mutations is significantly stable, e.g., due to the presence of stabilizing V69A and Q74P mutations.
  • V69A and Q74P substitutions do not substantially increase the binding affinity of the IL-2 variant for CD25, but rather stabilize the IL-2 variant in an active conformation sufficient for binding to CD25. Therefore, an IL-2 variant comprising these mutations selectively activates regulatory T cells (Treg) and is significantly stable.
  • an IL-2 variant comprising the aforesaid mutations has reduced or decreased binding capacity and/or binding affinity for CD25, which improves the lifetime of the IL-2 variant.
  • an IL-2 variant comprising these mutations does not substantially promote expansion, activation, survival, and/or proliferation of T effector cells and/or natural killer (NK) cells in vitro and/or in vivo.
  • NK natural killer
  • an IL-2 variant comprising the aforesaid mutations has reduced incorrect disulfide pairing and improved stability, e.g., due to the presence of the C125S mutation.
  • an IL-2 agent comprising the H16L mutation has reduced binding affinity for CD 122 and/or CD 132 and/or increased potency and selectivity for Treg over other T cell types, compared to an IL-2 agent comprising other H16 mutations. These properties make an IL-2 variant comprising these mutations particularly suitable for treating disorders and conditions arising from abnormal immune responses.
  • an IL-2 variant (e.g., IL-2 variant or IL-2 fusion protein) comprising an amino acid substitution at position Hl 6 in combination with amino acid substitutions at positions V69, Q74, and C125 (e.g., H16L, V69A, Q74P, and C125S), has inter alia one or more (e.g., 2, 3, 4, 5, 6, 7, or all) of the following properties relative to a wild-type IL-2 or a reference IL-2 variant that does not comprise the amino acid substitutions: (i) enhanced or increased stability in vitro or in vivo', (ii) reduced or decreased binding capacity and/or binding affinity for human CD 122 in vitro and/or in vivo', (iii) reduced or decreased binding capacity and/or binding affinity for human CD132 in vitro and/or in vivo-, (iv) reduced or decreased affinity of the IL-2 variant for the heterodimeric IL-2 receptor composed of human CD122 and human CD132 (i.e.
  • human CD122/CD132 heterodimer in vitro and/or in vivo; (v) reduced or decreased binding capacity and/or binding affinity for human CD25 in vitro and/or in vivo; (vi) selective binding to regulatory T cells (e.g. Foxp3 + T cells); (vii) selective activation of the IL-2 signaling pathway in T regulatory cells (Tregs) in vitro or in vivo; or (viii) enhanced or increased ability to induce or promote Treg expansion, activity, survival and/or proliferation.
  • regulatory T cells e.g. Foxp3 + T cells
  • the IL-2 variant comprises, or consists of, an amino acid sequence chosen from: SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 35
  • the IL-2 variant comprises, or consists of, the amino acid sequence of SEQ ID NO: 4, or an amino acid sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 amino acids thereto.
  • the IL-2 variant comprises, or consists of, the amino acid sequence of SEQ ID NO: 5, or an amino acid sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 amino acids thereto.
  • the IL-2 variant comprises, or consists of, the amino acid sequence of SEQ ID NO: 11, or an amino acid sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 amino acids thereto.
  • the IL-2 variant comprises, or consists of, the amino acid sequence of SEQ ID NO: 1000, or an amino acid sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 amino acids thereto.
  • the IL-2 variant comprises, or consists of, the amino acid sequence of SEQ ID NO: 1001, or an amino acid sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 amino acids thereto.
  • the IL-2 variant comprises, or consists of, the amino acid sequence of SEQ ID NO: 1002, or an amino acid sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 amino acids thereto.
  • the IL-2 variant comprises, or consists of, the amino acid sequence of any of SEQ ID NOs: 4, 5, 11, 1000, 1001, or 1002, or a functional fragment thereof. In an embodiment, the IL-2 variant comprises, or consists of, the amino acid sequence of SEQ ID NO: 4 or 5, or a functional fragment thereof. In an embodiment, the IL-2 variant comprises, or consists of, the amino acid sequence of SEQ ID NO: 4, or a functional fragment thereof. In an embodiment, the IL-2 variant comprises, or consists of, the amino acid sequence of SEQ ID NO: 5, or a functional fragment thereof. In an embodiment, the IL-2 variant comprises, or consists of, the amino acid sequence of SEQ ID NO: 11, or a functional fragment thereof.
  • the IL-2 variant comprises, or consists of, the amino acid sequence of SEQ ID NO: 1000, or a functional fragment thereof. In an embodiment, the IL-2 variant comprises, or consists of, the amino acid sequence of SEQ ID NO: 1001, or a functional fragment thereof. In an embodiment, the IL-2 variant comprises, or consists of, the amino acid sequence of SEQ ID NO: 1002, or a functional fragment thereof.
  • an IL-2 variant comprising, or consisting of, the amino acid sequence of SEQ ID NO: 5, or a functional fragment thereof, can have at least one or more of the following advantageous properties: (i) has reduced binding affinity for CD 122 and/or CD 132, which increases the potency and selectivity of the IL-2 agent for regulatory T cells (Treg) compared to other T cell types; (ii) is significantly stable, e.g, due to the presence of stabilizing V69A and Q74P mutations; (iii) has reduced or decreased binding capacity and/or binding affinity for CD25, which improves the lifetime of the IL-2 agent; (iv) does not substantially promote expansion, activation, survival, and/or proliferation of T effector cells and/or natural killer (NK) cells in vitro and/or in vivo', and/or (v) has reduced incorrect disulfide pairing and improved stability, e.g., due to the presence of the C125S mutation.
  • an IL-2 agent comprising the H16L mutation has reduced binding affinity for CD 122 and/or CD 132 and/or increased potency and selectivity for Treg over other T cell types, compared to an IL-2 agent comprising other H16 mutations.
  • an IL-2 variant comprising, or consisting of, the amino acid sequence SEQ ID NO: 5, or a functional fragment thereof, or an amino acid sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 amino acids thereto, has inter alia one or more (e.g., 2, 3, 4, 5, 6, 7, or all) of the following properties relative to a wild-type IL-2 or a reference IL-2 variant that does not comprise the amino acid substitutions: (i) enhanced or increased stability in vitro or in vivo', (ii) reduced or decreased binding capacity and/or binding affinity for human CD 122 in vitro and/or in vivo', (iii) reduced or decreased binding capacity and/or binding affinity for human CD 132 in vitro and/or in vivo; (iv) reduced or
  • human CD122/CD132 heterodimer in vitro and/or in vivo; (v) reduced or decreased or substantially unchanged binding capacity and/or binding affinity for human CD25 in vitro and/or in vivo; (vi) selective binding to regulatory T cells (e.g. Foxp3 + T cells); (vii) selective activation of the IL-2 signaling pathway in T regulatory cells (Tregs) in vitro or in vivo; or (viii) enhanced or increased ability to induce or promote Treg expansion, activity, survival and/or proliferation.
  • regulatory T cells e.g. Foxp3 + T cells
  • the disclosure provides IL-2 fusion proteins, IL-2 complexes, and IL-2 conjugates comprising an IL-2 variant described herein.
  • one or more different and/or improved properties ascribed to an IL-2 variant described herein is maintained, transferred, or imparted to the IL-2 fusion protein, IL-2 complex, or IL-2.
  • the terms “IL-2 variant” and “IL-2 mutein” may be used interchangeably herein.
  • the IL-2 variant comprises a polypeptide (sometime referred to herein as “IL-2 variant polypeptide”).
  • IL-2 variant polypeptide This disclosure provides an isolated nucleic acid molecule encoding an IL-2 variant described herein, and vectors and host cells thereof.
  • the nucleic acid molecule includes, but is not limited to, RNA, genomic DNA and cDNA.
  • the IL-2 agent comprises an IL-2 fusion protein, e.g. , an IL-2 fusion protein described herein.
  • the IL-2 fusion protein comprises an IL-2 variant, e.g, an IL-2 variant described herein.
  • the IL-2 fusion protein comprises one or more amino acid alterations (e.g., substitutions) described in Table 9.
  • the IL-2 fusion protein comprises an amino acid sequence described in Table 9, or a functional fragment thereof.
  • the IL-2 variant is encoded by a nucleic acid comprising a nucleotide sequence described herein, e.g., in Table 10.
  • the IL-2 fusion proteins described herein which have reduced human CD25 and/or reduced human CD122/CD132 binding affinity relative to a IL-2 fusion protein comprising a wild-type human IL-2 or a reference IL- 2 fusion protein, can have improved potency and/or selectivity for binding to and activating regulatory T cells (Tregs) than IL-2 fusion proteins comprising a wild-type human IL-2 or other IL-2 fusion protein.
  • Tregs regulatory T cells
  • the IL-2 fusion protein has one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more) properties described herein, e.g., different and/or improved properties, relative to an IL-2 fusion protein comprising a wild-type IL-2 or a reference IL-2 fusion protein.
  • the IL-2 fusion protein comprises one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) amino acid alterations (e.g., substitutions) that provide different and/or improved properties, relative to an IL-2 fusion protein comprising a wild-type IL-2 or a reference IL-2 fusion protein.
  • the IL-2 fusion protein has one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or all) of the following different and/or improved properties (e.g., as determined by an assay described herein), relative to an IL-2 fusion protein comprising a wild-type IL-2 or a reference IL-2 fusion protein: i) altered (e.g., enhanced or increased) expression in vitro and/or in vivo', ii) altered (e.g., reduced or decreased) aggregation in vitro and/or in vivo-, iii) altered (e.g., enhanced or increased) stability in vitro and/or in vivo', iv) altered (e.g., enhanced or increased) half-life in vitro and/or in vivo', v) altered (e.g., reduced or decreased) turnover and/or clearance in vivo-, vi) altered (e.g., reduced or decreased) susceptibility to proteolysis in vitro
  • the IL-2 fusion protein has altered (e.g., enhanced or increased) expression in vitro and/or in vivo, relative to an IL-2 fusion protein comprising a wild-type IL-2 or a reference IL-2 fusion protein.
  • the IL-2 fusion protein has enhanced or increased expression (e.g., in a bacterial or mammalian cell) relative to an IL-2 fusion protein comprising a wild-type IL-2.
  • the IL-2 fusion protein has enhanced or increased expression (e.g., in bacterial or mammalian cell) relative to a reference IL-2 fusion protein.
  • the expression of the IL-2 fusion protein is increased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more. In an embodiment, the expression of the IL-2 fusion protein is increased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9- fold, or about 10-fold, or more.
  • the IL-2 fusion protein expresses at a higher or increased level in vitro and/or in vivo, e.g., increased by about 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100% or more e.g., relative to an IL-2 fusion protein comprising a wildtype IL-2 or a reference IL-2 fusion protein e.g, as determined by an assay of protein concentration.
  • the IL-2 fusion protein expresses at a higher or increased level, e.g., increased by about 0.5-fold, about 1-fold, about 1.5-fold, about 2-fold, about 2.5-fold, about 3-fold, about 3.5-fold, about 4-fold, about 4.5-fold, about 5-fold, about 5.5-fold, about 6-fold, about 6.5-fold, about 7-fold, about 7.5-fold, about 8-fold, about 8.5-fold, about 9-fold, about 9.5-fold, about 10-fold or more e.g, relative to an IL-2 fusion protein comprising a wild-type IL-2 or a reference IL-2 fusion protein e.g, as determined by an assay of protein concentration.
  • the IL-2 fusion protein has altered (e.g., reduced or decreased) aggregation in vitro and/or in vivo, relative to an IL-2 fusion protein comprising a wild-type IL-2 or a reference IL-2 fusion protein.
  • the IL-2 fusion protein has reduced or decreased aggregation relative to a wild type IL-2.
  • the IL-2 fusion protein has reduced or decreased aggregation relative to a reference IL-2 fusion protein.
  • the aggregation of the IL-2 fusion protein is decreased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more.
  • the aggregation of the IL-2 fusion protein is decreased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or about 10-fold, or more.
  • the IL-2 fusion protein aggregates at lower or decreased level in vitro and/or in vivo, e.g., decreased by about 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100% or more e.g., relative to an IL-2 fusion protein comprising a wild-type IL-2 or a reference IL-2 fusion protein e.g, as determined by melting temperature analysis (e.g., using fluorimetry), dynamic light scattering, and/or size-exclusion chromatography.
  • melting temperature analysis e.g., using fluorimetry
  • dynamic light scattering e.g., dynamic light scattering
  • size-exclusion chromatography e.g., as determined by melting temperature analysis (e.g., using fluorimetry
  • the IL-2 fusion protein aggregates at lower or decreased level, e.g, decreased by about 0.5-fold, about 1- fold, about 1.5-fold, about 2-fold, about 2.5-fold, about 3-fold, about 3.5-fold, about 4-fold, about 4.5- fold, about 5-fold, about 5.5-fold, about 6-fold, about 6.5-fold, about 7-fold, about 7.5-fold, about 8- fold, about 8.5 -fold, about 9-fold, about 9.5 -fold, about 10-fold or more e.g, relative to an IL-2 fusion protein comprising a wild-type IL-2 or a reference IL-2 fusion protein e.g, as determined by melting temperature analysis (e.g., using fluorimetry), dynamic light scattering, and/or size -exclusion chromatography.
  • melting temperature analysis e.g., using fluorimetry
  • dynamic light scattering e.g., dynamic light scattering
  • size -exclusion chromatography e.g., as determined by melting temperature analysis (e
  • the IL-2 fusion protein has altered (e.g., enhanced or increased) stability in vitro and/or in vivo, relative to an IL-2 fusion protein comprising a wild-type IL-2 or a reference IL-2 fusion protein.
  • the IL-2 fusion protein has enhanced or increased stability relative to an IL-2 fusion protein comprising a wild-type IL-2.
  • the IL-2 fusion protein has enhanced or increased stability relative to a reference IL-2 fusion protein.
  • the stability of the IL-2 fusion protein is increased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more.
  • the stability of the IL-2 fusion protein is increased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9- fold, or about 10-fold, or more.
  • the IL-2 fusion protein has enhanced or increased stability in vitro and/or in vivo, e.g., increased by about 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100% or more, or e.g., increased by about 0.5-fold, about 1-fold, about 1.5- fold, about 2-fold, about 2.5-fold, about 3-fold, about 3.5-fold, about 4-fold, about 4.5-fold, about 5- fold, about 5.5-fold, about 6-fold, about 6.5-fold
  • the IL-2 fusion protein has altered (e.g., enhanced or increased) half-life in vitro and/or in vivo, relative to an IL-2 fusion protein comprising a wild-type IL-2 or a reference IL-2 fusion protein.
  • the IL-2 fusion protein has enhanced or increased half-life relative to an IL-2 fusion protein comprising a wild-type IL-2.
  • the IL-2 fusion protein has enhanced or increased half-life relative to a reference IL-2 fusion protein.
  • the halflife of the IL-2 fusion protein is increased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more.
  • the half-life of the IL-2 fusion protein is increased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or about 10-fold, or more.
  • the IL-2 fusion protein has enhanced or increased half-life in vitro and/or in vivo, e.g, increased by about 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100% or more, or e.g., greater than about 0.5-fold, about 1-fold, about 1.5-fold, about 2- fold, about 2.5-fold, about 3-fold, about 3.5-fold, about 4-fold, about 4.5-fold, about 5-fold, about 5.5- fold, about 6-fold, about
  • the IL-2 fusion protein has altered (e.g., reduced or decreased) turnover in vitro and/or in vivo, relative to an IL-2 fusion protein comprising a wild-type IL-2 or a reference IL-2 fusion protein.
  • the IL-2 fusion protein has reduced or decreased turnover relative to an IL-2 fusion protein comprising a wild-type IL-2.
  • the IL-2 fusion protein has reduced or decreased turnover relative to a reference IL-2 fusion protein.
  • the turnover of the IL-2 fusion protein is decreased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more.
  • the turnover of the IL-2 fusion protein is decreased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9- fold, about 10-fold, or more.
  • the IL-2 fusion protein has a lower, reduced or decreased rate or level of turnover and/or clearance in vivo, e.g., decreased by about 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100% or more, or e.g., decreased by about 0.5- fold, about 1-fold, about 1.5-fold, about 2-fold, about 2.5-fold, about 3-fold, about 3.5-fold, about 4- fold, about 4.5-fold, about 5-fold, about 5.5-fold, about 6-fold,
  • the IL-2 fusion protein provided by the disclosure comprise the property of having altered (e.g., reduced or decreased) susceptibility to proteolysis in vitro and/or in vivo, relative to an IL-2 fusion protein comprising a wild-type IL-2 or a reference IL-2 fusion protein.
  • the IL-2 fusion protein has reduced or decreased susceptibility to proteolysis relative to IL-2 (e.g., wild type human IL-2).
  • the IL-2 fusion protein has reduced or decreased susceptibility to proteolysis relative to a reference IL-2 fusion protein.
  • the susceptibility to proteolysis of the IL-2 fusion protein is decreased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more. In an embodiment, the susceptibility to proteolysis of the IL-2 fusion protein is decreased by about 0.5-fold, 1-fold, 2-fold, 3- fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or about 10-fold, or more.
  • the IL-2 fusion protein has altered (e.g., enhanced or increased) resistance to proteolysis in vitro and/or in vivo, relative to an IL-2 fusion protein comprising a wild-type IL-2 or a reference IL-2 fusion protein.
  • the IL-2 fusion protein has enhanced or increased resistance to proteolysis relative to an IL-2 fusion protein comprising a wild-type IL-2.
  • the IL-2 fusion protein has enhanced or increased resistance to proteolysis relative to a reference IL-2 fusion protein.
  • the resistance to proteolysis of the IL-2 fusion protein is increased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more. In an embodiment, the resistance to proteolysis of the IL-2 fusion protein is increased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or about 10-fold, or more.
  • the IL-2 fusion protein has altered (e.g., reduced or decreased) binding capacity and/or binding affinity for human CD25 in vitro and/or in vivo, relative to an IL-2 fusion protein comprising a wild-type IL-2 or a reference IL-2 fusion protein.
  • the IL-2 fusion protein has reduced or decreased binding capacity and/or binding affinity for human CD25 relative to a wild-type human IL-2).
  • the IL-2 fusion protein has reduced or decreased binding capacity and/or binding affinity for human CD25 relative to a reference IL-2 fusion protein.
  • the binding capacity and/or binding affinity of the IL-2 fusion protein for human CD25 is decreased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more. In an embodiment, the binding capacity and/or binding affinity of the IL-2 fusion protein for human CD25 is decreased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5- fold, 6-fold, 7-fold, 8-fold, 9-fold, or about 10-fold, or more.
  • the IL-2 fusion protein has reduced or decreased binding affinity for CD25 (e.g., human CD25), e.g., decreased by about 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100% or more, or e.g., decreased by about 0.5-fold, about 1-fold, about 1.5-fold, about 2-fold, about 2.5-fold, about 3-fold, about 3.5-fold, about 4-fold, about 4.5-fold, about 5-fold, about 5.5-fold, about 6-fold, about 6.5-fold, about 7-fold, about 7.5-fold, about 8-fold, about 8.5-fold, about 9-fold, about 9.5-fold, about 10-fold or more e.g.
  • an IL-2 fusion protein comprising a wild-type IL-2 or a reference IL-2 fusion protein e.g., as determined by yeast surface display, surface plasmon resonance (e.g. Biacore) and/or bio-layer interferometry (e.g. Octet binding).
  • the IL-2 fusion protein binds to CD25 (e.g. , human CD25) with low affinity, e.g., with a dissociation constant (K D ) of about 5-500 pM, e.g., about 5, about 10, about 15, about 20, about 25, about 30, about 35, about 40, about 45, about 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 105, about 110, about 115, about 120, about 125, about 130, about 135, about 140, about 145, about 150, about 200, about 250, about 300, about 350, about 400, about 450, or about 500 pM, or e.g., about 10 to about 400 pM, about 20 to about 300 pM, about 50 to about 200 pM, about 100 to about 150 pM, about 5 to about 10 pM, about 10 to about 20 pM, about 20 to about 30 pM, or about 30 to about 40
  • K D
  • the IL-2 fusion protein binds to CD25 (e.g. , human CD25) with low affinity, e.g., with a dissociation constant (K D ) of about 0.1-10 nM, e.g., about 0.1, about 0.2, about 0.3, about 0.4, about 0.5, about 0.6, about 0.7, about 0.8, about 0.9, about 1, about 1.5, about 2, about 2.5, about 3, about 3.5, about 4, about 4.5, about 5, about 6, about 7, about 8, about 9, or about 10 nM, or e.g., about 0.2 to about 5 nM, about 0.5 to about 2 nM, about 1 to 1.5 nM, about 0.1 to about 0.2 nM, about 0.2 to about 0.3 nM, about 0.3 to about 0.4 nM, or about 0.4 to about 0.5 nM, e.g., about 0.5 to about 0.6 nM, about 0.6 to about 0.7 nM, about 0.7 to about 0.8
  • nM e.g, as determined by surface plasmon resonance (e.g. Biacore) and/or bio-layer interferometry (e.g., Octet binding).
  • surface plasmon resonance e.g. Biacore
  • bio-layer interferometry e.g., Octet binding
  • the IL-2 fusion protein has altered (e.g., reduced or decreased) binding capacity and/or binding affinity for human CD 132 in vitro and/or in vivo, relative to an IL-2 fusion protein comprising a wild-type IL-2 or a reference IL-2 fusion protein. In an embodiment, the IL-2 fusion protein has reduced or decreased binding capacity and/or binding affinity for human CD 132 relative to an IL-2 fusion protein comprising a wild-type IL-2. In an embodiment, the IL-2 fusion protein has reduced or decreased binding capacity and/or binding affinity for human CD132 relative to a reference IL-2 fusion protein.
  • the binding capacity and/or binding affinity of the IL-2 fusion protein for human CD 132 is decreased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more. In an embodiment, the binding capacity and/or binding affinity of the IL-2 fusion protein for human CD132 is decreased by about 0.5-fold, 1-fold, 2- fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or about 10-fold, or more.
  • the IL-2 fusion protein has altered (e.g., reduced or decreased) binding capacity and/or binding affinity for the human dimeric IL-2 receptor comprising human CD 122 and human CD132 in vitro and/or in vivo, relative to an IL-2 fusion protein comprising a wild-type IL-2 or a reference IL-2 fusion protein.
  • the IL-2 fusion protein has reduced or decreased binding capacity and/or binding affinity for the human dimeric IL-2 receptor comprising human CD122 and human CD132 relative to an IL-2 fusion protein comprising a wild-type IL-2.
  • the IL-2 fusion protein has reduced or decreased binding capacity and/or binding affinity for the human dimeric IL-2 receptor comprising human CD 122 and human CD 132 relative to a reference IL-2 fusion protein.
  • the binding capacity and/or binding affinity of the IL-2 fusion protein for the human dimeric IL-2 receptor comprising human CD 122 and human CD 132 is decreased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more.
  • the binding capacity and/or binding affinity of the IL-2 fusion protein for the human dimeric IL-2 receptor comprising human CD122 and human CD132 is decreased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or about 10-fold, or more.
  • the IL-2 fusion protein has altered (e.g., enhanced, increased, and/or selective) binding to Tregs in vitro and/or in vivo, relative to an IL-2 fusion protein comprising a wildtype IL-2 or a reference IL-2 fusion protein.
  • the IL-2 fusion protein has enhanced or increased binding to Tregs relative to an IL-2 fusion protein comprising a wild-type IL-2.
  • the IL-2 fusion protein has selective binding to Tregs relative to IL-2 (e.g., wild type human IL-2).
  • the IL-2 fusion protein has enhanced or increased binding to Tregs relative to a reference IL-2 fusion protein.
  • the IL-2 fusion protein has selective binding to Tregs relative to a reference IL-2 fusion protein.
  • the binding to Tregs is increased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more.
  • the binding to Tregs is increased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or about 10-fold, or more.
  • the IL-2 fusion protein has reduced or decreased binding affinity for CD122/CD132 heterodimer (e.g., human CD122/CD132 heterodimer), e.g., decreased by about 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100% or more, or e.g., decreased by about 0.5-fold, about 1-fold, about 1.5-fold, about 2-fold, about 2.5-fold, about 3-fold, about 3.5-fold, about 4-fold, about 4.5-fold, about 5-fold, about 5.5-fold, about 6-fold, about 6.5-fold, about 7-fold, about 7.5-fold, about 8-fold, about 8.5-fold, about 9-fold, about 9.5-fold, about 10-fold or more e.g.
  • the IL-2 fusion protein binds to CD122/CD132 heterodimer (e.g., human CD122/CD132 heterodimer) with low affinity, e.g., with a dissociation constant (K D ) of about 0.2-20 nM, e.g., about 0.2, about 0.3, about 0.4, about 0.5, about 0.6, about 0.7, about 0.8, about 0.9, about 1, about 1.1, about 1.2, about 1.3, about 1.4.
  • K D dissociation constant
  • nM about 1.5, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, or about 20 nM, or e.g., about 0.5 to about 15 nM, about 1 to about 10 nM, about 2 to about 5 nM, about 0.2 to about 0.3 nM, about 0.3 to about 0.4 nM, about 0.4 to about 0.5 nM, about 0.5 to about 0.6 nM, about 0.6 to about 0.7 nM, about 0.7 to about 0.8 nM, about 0.8 to about 0.9 nM, about 0.9 to about 1 nM, about 1 to about 1.1 nM, about 1.1 to about 1.2 nM, about 1.2 to about 1.3 nM, about 1.3 to about 1.4 nM, about 1.4 to about 1.5 nM, about 1.5 to about 2 nM, about 2 to about 3 nM, about 3 to about 4 nM, about 4 to about 5
  • the IL-2 fusion protein binds to CD122/CD132 heterodimer (e.g., human CD122/CD132 heterodimer) with low affinity, e.g., with a dissociation constant (K D ) of about 0.2-300 nM , e.g., about 0.2 nM, about 0.5 nM, about 1 nM, about 2 nM, about 5 nM, about 10 nM, about 15 nM, about 20 nM, about 25 nM, about 30 nM, about 40 nM, about 50 nM, about 60 nM, about 70 nM, about 80 nM, about 90 nM, about 100 nM, about 110 nM, about 120 nM, about 130 nM, about 140 nM, about 150 nM, about 160 nM, about 170 nM, about 180 nM, about 190 nM, about 200 nM, about 210 nM, about 2
  • the IL-2 fusion protein has altered (e.g., enhanced, increased, and/or selective) binding to Tregs in vitro and/or in vivo, relative to an IL-2 fusion protein comprising wildtype IL-2 or a reference IL-2 fusion protein.
  • the IL-2 fusion protein has enhanced or increased binding to Tregs relative to an IL-2 fusion protein comprising wild-type IL-2.
  • the IL-2 fusion protein has selective binding to Tregs relative to IL-2 (e.g., wild type human IL-2).
  • the IL-2 fusion protein has enhanced or increased binding to Tregs relative to a reference IL-2 fusion protein.
  • the IL-2 fusion protein has selective binding to Tregs relative to a reference IL-2 fusion protein.
  • the binding to Tregs is increased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more.
  • the binding to Tregs is increased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or about 10-fold, or more.
  • the IL-2 fusion protein has altered (e.g., enhanced, increased, and/or selective) activation of the IL-2 signaling pathway in Tregs in vitro and/or in vivo, relative to an IL-2 fusion protein comprising a wild-type IL-2 or a reference IL-2 fusion protein.
  • the IL-2 fusion protein has enhanced or increased activation of the IL-2 signaling pathway in Tregs relative to an IL-2 fusion protein comprising a wild-type IL-2.
  • the IL-2 fusion protein has selective activation of the IL-2 signaling pathway in Tregs relative to an IL-2 fusion protein comprising a wild-type IL-2.
  • the IL-2 fusion protein has enhanced or increased activation of the IL-2 signaling pathway in Tregs relative to a reference IL-2 fusion protein. In an embodiment, the IL-2 fusion protein has selective activation of the IL-2 signaling pathway in Tregs relative to a reference IL-2 fusion protein. In an embodiment, the activation of the IL-2 signaling pathway in Tregs is increased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more.
  • the activation of the IL-2 signaling pathway in Tregs is increased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8- fold, 9-fold, or about 10-fold, or more.
  • the IL-2 fusion protein selectively activates IL-2 signaling in T regulatory cells in vitro and/or in vivo, e.g., having an T helper EC50/Treg EC50 ratio greater than about 1, about 2, about 3, about 4, about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1000, 1500, 2000, 2500, or about 3000 or more relative to an IL-2 fusion protein comprising a wild-type IL-2 or a reference IL-2 fusion protein e.g, as determined flow cytometry.
  • the IL-2 fusion protein selectively activates IL-2 signaling in T regulatory cells in vitro and/or in vivo, e.g., having an NK cell EC50/Treg EC50 ratio greater than e.g., about 1, about 2, about 3, about 4, about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1000, 1500, 2000, 2500, or about 3000 or more, or e.g., greater than 1 and about 1 to 2, about 2 to 3, about 3 to 4, about 4 to 5, greater than 1 and about 1 to 10, greater than 1 and about 1 to 20, greater than 1 and about 1 to 30, greater than 1 and about 1 to 40, greater than 1 and about 1 to 50, about 2 to 10, about 2 to 20, about 2 to 30, about 2 to 40, greater than
  • the IL-2 fusion protein has altered (e.g., enhanced, increased, and/or selective) ability to induce or promote Treg expansion, activity, survival, and/or proliferation in vitro and/or in vivo, relative to an IL-2 fusion protein comprising a wild-type IL-2 or a reference IL-2 fusion protein.
  • the IL-2 fusion protein has enhanced or increased ability to induce or promote Treg expansion, activity, survival, and/or proliferation relative to an IL-2 fusion protein comprising a wild-type IL-2.
  • the IL-2 fusion protein has selective ability to induce or promote Treg expansion, activity, survival, and/or proliferation relative to an IL-2 fusion protein comprising a wild-type IL-2.
  • the IL-2 fusion protein has enhanced or increased ability to induce or promote Treg expansion, activity, survival, and/or proliferation relative to a reference IL-2 fusion protein. In an embodiment, the IL-2 fusion protein has selective ability to induce or promote Treg expansion, activity, survival, and/or proliferation relative to a reference IL-2 fusion protein. In an embodiment, the ability to induce or promote Treg expansion, activity, survival, and/or proliferation is increased by about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or about 100%, or more.
  • the ability to induce or promote Treg expansion, activity, survival, and/or proliferation is increased by about 0.5-fold, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, or about 10-fold, or more.
  • the IL-2 fusion protein has enhanced or increased potency and/or ability to induce or promote T regulatory cell activity, e.g, having an EC50 for Tregs that is lower by about 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100% or more, or e.g., decreased by about 0.5-fold, about 1-fold, about 1.5-fold, about 2-fold, about 2.5-fold, about 3-fold, about 3.5-fold, about 4-fold, about 4.5-fold, about 5-fold, about 5.5-fold, about 6-fold, about 6.5-fold, about 7-fold, about 7.5-fold, about 8-fold, about 8.5-fold, about 9-fold, about 9.5-fold, about 10-fold or more e.g, relative to an EC50 for
  • the IL-2 fusion protein has reduced or decreased potency and/or ability to induce or promote T regulatory cell activity, e.g, having an EC50 for Tregs that is higher by about 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% or more, or e.g., decreased by about 0.5-fold, about 1-fold, about 1.5-fold, about 2-fold, about 2.5-fold, about 3-fold, about 3.5- fold, about 4-fold, about 4.5-fold, about 5-fold, about 5.5-fold, about 6-fold, about 6.5-fold, about 7- fold, about 7.5-fold, about 8-fold, about 8.5-fold, about 9-fold, about 9.5-fold, about 10-fold, about 50-fold, about 100-fold
  • the T helper cell described herein is a CD45+CD3+CD4+Foxp3- cell, e.g., determined by flow cytometry.
  • the Treg described herein is CD45+CD3+CD4+Foxp3+ cell, e.g., determined by flow cytometry.
  • the NK cell described herein is a CD45+CD3- cell that is CD56+ and/or CD16+, e.g., determined by flow cytometry.
  • the NK cell described herein is a CD45+CD3-CD56+ cell, e.g., determined by flow cytometry.
  • the IL-2 fusion protein has one or more of the same, or substantially the same, structural and/or functional properties, as an IL-2 fusion protein comprising a wild-type IL-2 or a reference IL-2 fusion protein.
  • the reference IL-2 fusion protein comprises an amino acid sequence that has about 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity to an IL-2 fusion protein described herein.
  • the reference IL-2 fusion protein comprises an IL-2 variant comprising the amino acid sequence of SEQ ID NO: 57.
  • the IL-2 fusion protein comprises an amino acid sequence that is at least 80%, 85%, 90%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO: 57 and comprises one or more (2, 3, 4, 5, 6, 7, 8, 9, 10, or more) amino acid alterations (e.g., substitutions) described herein.
  • the IL-2 fusion protein comprises an IL-2 polypeptide (e.g., a human IL-2 polypeptide) described herein.
  • the IL-2 fusion protein is encoded by a nucleic acid comprising a nucleotide sequence described herein.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or all) of positions in IL-2, as described herein.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g., substitution) at one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or all) of positions chosen from T3, H16, 128, K35, R38, F42, E68, V69, Q74, D84, S87, N88, 192, C125, or Q126 in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position T3 in IL-2. In an embodiment, the IL-2 fusion protein comprises an amino acid alteration (e.g., substitution) at position H16 in IL-2. In an embodiment, the IL-2 fusion protein comprises an amino acid alteration (e.g., substitution) at position 128 in IL-2. In an embodiment, the IL-2 fusion protein comprises an amino acid alteration (e.g., substitution) at position K35 in IL-2. In an embodiment, the IL-2 fusion protein comprises an amino acid alteration (e.g., substitution) at position R38 in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g., substitution) at position F42 in IL-2. In an embodiment, the IL-2 fusion protein comprises an amino acid alteration (e.g., substitution) at position E68 in IL-2. In an embodiment, the IL-2 fusion protein comprises an amino acid alteration (e.g., substitution) at position V69 in IL-2. In an embodiment, the IL-2 fusion protein comprises an amino acid alteration (e.g., substitution) at position Q74 in IL-2. In an embodiment, the IL-2 fusion protein comprises an amino acid alteration (e.g., substitution) at position D84 in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g., substitution) at position S87 in IL-2. In an embodiment, the IL-2 fusion protein comprises an amino acid alteration (e.g., substitution) at position N88 in IL-2. In an embodiment, the IL-2 fusion protein comprises an amino acid alteration (e.g., substitution) at position 192 in IL-2. In an embodiment, the IL-2 fusion protein comprises an amino acid alteration (e.g., substitution) at position C125 in IL-2. In an embodiment, the IL-2 fusion protein comprises an amino acid alteration (e.g., substitution) at position Q126 in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position V69, Q74, or both, in IL-2. In an embodiment, the IL-2 fusion protein comprises an amino acid alteration (e.g., substitution) at positions V69 and Q74 in IL-2. In an embodiment, the IL-2 fusion protein comprises the amino acid substitution V69A in IL-2. In an embodiment, the IL-2 fusion protein comprises the amino acid substitution Q74P in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position H16, 192, D84, or a combination thereof, in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g., substitution) at position H16, optionally wherein the amino acid substitution is H16N, H16L, or H16D, in IL-2.
  • the IL-2 fusion protein comprises the amino acid substitution H16N in IL-2.
  • the IL-2 fusion protein comprises the amino acid substitution H16L in IL-2.
  • the IL-2 fusion protein comprises the amino acid substitution H16D in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position at 192, optionally wherein the amino acid substitution is I92S, in IL-2. In an embodiment, the IL-2 fusion protein comprises the amino acid substitution I92S in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position D84, optionally wherein the amino acid substitution is D84V, in IL-2. In an embodiment, the IL-2 fusion protein comprises the amino acid substitution is D84V in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position K35, R38, F42, E68, or a combination thereof, in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g., substitution) at position K35, optionally wherein the amino acid substitution is K35E, in IL-2.
  • IL-2 fusion protein comprises the amino acid substitution K35E in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position R38, optionally wherein the amino acid substitution is R38E, R38N or R38Q, in IL-2.
  • the IL-2 fusion protein comprises the amino acid substitution R38N in IL- 2.
  • the IL-2 fusion protein comprises the amino acid substitution R38Q in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position F42, optionally wherein the amino acid substitution is F42K or F42Q, in IL-2. In an embodiment, the IL-2 fusion protein comprises the amino acid substitution F42K in IL-2. In an embodiment, the IL-2 fusion protein comprises the amino acid substitution F42Q in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution): (i) at (a) positions V69 and Q74, (b) position K35, or (c) positions V69, Q74, and K35; and (ii) at one, two, or all of positions H16, 192, or D84, in IL-2.
  • the IL-2 fusion protein further comprises an amino acid alteration (e.g., substitution) at one, two, or all of positions R38, F42, or E68, in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution): (i) at (a) positions V69 and Q74, (b) position K35, or (c) positions V69, Q74, and K35; and (ii) at (a) one, two, or all of positions H16, 192, or D84; or (b) one, two, or all of positions R38, F42, or E68, in IL-2.
  • amino acid alteration e.g. , substitution
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution): (i) at (a) positions V69 and Q74, (b) position K35, or (c) positions V69, Q74, and K35; and (ii) at (a) one, two, or all of positions H16, 192, or D84; and (b) one, two, or all of positions R38, F42, or E68, in IL-2.
  • amino acid alteration e.g. , substitution
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position V69, Q74, and H16, optionally wherein the amino acid substitution is V69A, Q74P, and H16N or H16L, respectively, in IL-2.
  • the IL-2 fusion protein comprises the amino acid substitutions V69A, Q74P, and H16N or H16L, in IL-2.
  • the IL-2 fusion protein comprises the amino acid substitutions V69A, Q74P, and H16N, in IL-2.
  • the IL-2 fusion protein comprises the amino acid substitutions V69A, Q74P, and H16L, in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position V69, Q74, and 192, optionally wherein the amino acid substitution is V69A, Q74P, and I92S, respectively, in IL-2.
  • the IL-2 fusion protein comprises the amino acid substitutions V69A, Q74P, and I92S, in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position V69, Q74, and D84, optionally wherein the amino acid substitution is V69A, Q74P, and D84V, respectively, in IL-2.
  • the IL-2 fusion protein comprises the amino acid substitutions V69A, Q74P, and D84V, in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position V69, Q74, and R38, optionally wherein the amino acid substitution is V69A, Q74P, and R38Q, respectively, in IL-2.
  • the IL-2 fusion protein comprises the amino acid substitutions V69A, Q74P, and R38Q, in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position V69, Q74, and F42, optionally wherein the amino acid substitution is V69A, Q74P, and F42Q, respectively, in IL-2.
  • the IL-2 fusion protein comprises the amino acid substitutions V69A, Q74P, and F42Q, in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position V69, Q74, and R38, optionally wherein the amino acid substitution is V69A, Q74P, and R38N, respectively, in IL-2.
  • the IL-2 fusion protein comprises the amino acid substitutions V69A, Q74P, and R38N, in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position V69, Q74, and R38, optionally wherein the amino acid substitution is V69A, Q74P, and R38E, respectively, in IL-2.
  • the IL-2 fusion protein comprises the amino acid substitution V69A, Q74P, and R38E, in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position V69, Q74, K35, and H16, optionally wherein the amino acid substitution is V69A, Q74P, K35E, and H16N, respectively, in IL-2.
  • the IL-2 fusion protein comprises the amino acid substitutions V69A, Q74P, K35E, and H16N, in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position V69, Q74, K35, H16, and R38, optionally wherein the amino acid substitution is V69A, Q74P, K35E, H16N, and R38N, respectively, in IL-2.
  • the IL-2 fusion protein comprises the amino acid substitutions V69A, Q74P, K35E, H16N, and R38N, in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position V69, Q74, H16, and R38, optionally wherein the amino acid substitution is V69A, Q74P, H16N, and R38N or R38Q, respectively, in IL-2.
  • the IL-2 fusion protein comprises the amino acid substitutions V69A, Q74P, H16N, and R38N or R38Q, in IL-2.
  • the IL-2 fusion protein comprises the amino acid substitutions V69A, Q74P, H16N, and R38N, in IL-2.
  • the IL-2 fusion protein comprises the amino acid substitutions V69A, Q74P, H16N, and R38Q, in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position 128, E68, S87, N88, QI 26, or a combination thereof, in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position 128, optionally wherein the amino acid substitution is I28T or I28F, in IL-2. In an embodiment, the IL-2 fusion protein comprises the amino acid substitution I28T in IL-2. In an embodiment, the IL-2 fusion protein comprises the amino acid substitution I28F in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position E68, optionally wherein the amino acid substitution is E68Q or E68N, in IL- 2. In an embodiment, the IL-2 fusion protein comprises the amino acid substitution E68Q in IL-2. In an embodiment, the IL-2 fusion protein comprises the amino acid substitution E68N in IL-2. In an embodiment, the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position S87, optionally wherein the amino acid substitution is S87R, in IL-2. In an embodiment, the IL-2 fusion protein comprises the amino acid substitution S87R in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position N88, optionally wherein the amino acid substitution is N88S, N88L, or N88D, in IL-2. In an embodiment, the IL-2 fusion protein comprises the amino acid substitution N88S, N88L, or N88D, in IL-2. In an embodiment, the IL-2 fusion protein comprises the amino acid substitution N88S in IL-2. In an embodiment, the IL-2 fusion protein comprises the amino acid substitution N88L in IL-2. In an embodiment, the IL-2 fusion protein comprises the amino acid substitution N88D in IL-2.
  • substitution e.g. , substitution
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position Q126, optionally wherein the amino acid substitution is Q126T, Q126K, or Q126R, in IL-2. In an embodiment, the IL-2 fusion protein comprises the amino acid substitution Q126T, Q126K, or Q126R, in IL-2. In an embodiment, the IL-2 fusion protein comprises the amino acid substitution Q126T, Q126K, or Q126R, in IL-2. In an embodiment, the IL-2 fusion protein comprises the amino acid substitution Q126T in IL-2. In an embodiment, the IL-2 fusion protein comprises the amino acid substitution Q126K in IL-2. In an embodiment, the IL-2 fusion protein comprises the amino acid substitution Q126R in IL-2.
  • substitution e.g. , substitution
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position C125 in IL-2, optionally wherein the amino acid substitution is C125S. In an embodiment, the IL-2 fusion protein comprises the amino acid substitution C125S in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position T3 in IL-2, optionally wherein the amino acid substitution is T3A. In an embodiment, the IL-2 fusion protein comprises the amino acid substitution T3A in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position V69, Q74, and C125, in IL-2, optionally wherein the amino acid substitution is V69A, Q74P, and C125S, respectively.
  • the IL-2 fusion protein comprises the amino acid substitutions V69A, Q74P, and C125S, in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position T3, H16, 192, in IL-2, or a combination thereof, optionally wherein the amino acid substitution is T3A, H16N, and I92S, in IL-2, respectively.
  • amino acid alteration e.g. , substitution
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position H16, V69, Q74, and C125, in IL-2, optionally wherein the amino acid substitution is H16N, V69A, Q74P, and C125S, in IL-2, respectively.
  • the IL-2 fusion protein comprises the amino acid substitutions H16N, V69A, Q74P, and C125S in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position H16, V69, Q74, and C125, in IL-2, optionally wherein the amino acid substitution is H16L, V69A, Q74P, and C125S, in IL-2, respectively.
  • the IL-2 fusion protein comprises the amino acid substitutions H16L, V69A, Q74P, and C125S, in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position H16, V69, Q74, 192, and C125, in IL-2, optionally wherein the amino acid substitution is H16L, V69A, Q74P, I92S, and C125S, in IL-2, respectively.
  • the IL-2 fusion protein comprises the amino acid substitutions H16L, V69A, Q74P, I92S, and C125S, in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position T3, V69, Q74, and C125, in IL-2, optionally wherein the amino acid substitution is T3A, V69A, Q74P, and C125S, in IL-2, respectively.
  • the IL-2 fusion protein comprises the amino acid substitutions T3A, V69A, Q74P, and C125S, in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position T3, H16, V69, Q74, and C125, in IL-2, optionally wherein the amino acid substitution is T3A, H16N or H16L, V69A, Q74P, and C125S, in IL-2, respectively.
  • the IL-2 fusion protein comprises the amino acid substitutions T3A, H16N, V69A, Q74P, and C125S.
  • the IL-2 fusion protein comprises the amino acid substitutions T3A, H16L, V69A, Q74P, and C125S, in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position T3, V69, Q74, 192, and C125, in IL-2, optionally wherein the amino acid substitution is T3A, V69A, Q74P, I92S, and C125S, in IL-2, respectively.
  • the IL- 2 fusion protein comprises the amino acid substitutions T3A, V69A, Q74P, I92S, and C125S, in IL-2.
  • the IL-2 fusion protein comprises the amino acid substitutions T3A, V69A, Q74P, I92S, and C125S, in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position H16, K35, V69 and Q74, optionally wherein the amino acid substitution is H16L, K35E, V69A, and Q74P, respectively, in IL-2.
  • the IL-2 fusion protein comprises the amino acid substitutions H16L, K35E, V69A, and Q74P, in IL-2.
  • the IL-2 fusion protein comprises an amino acid alteration (e.g. , substitution) at position H16, R38, V69A, and Q74P, optionally wherein the amino acid substitution is H16L, R38Q, V69A, and Q74P, respectively, in IL-2.
  • the IL-2 fusion protein comprises the amino acid substitutions H16L, R38Q, V69A, and Q74P, in IL-2.
  • the IL-2 fusion protein comprises the amino acid substitutions H16L, V69A, Q74P, and C125S, in IL-2.
  • an IL-2 fusion protein comprising the amino acid substitutions H16L, V69A, Q74P, and C125S, can have at least one or more of the following advantageous properties: (i) has reduced binding affinity for CD122 and/or CD132, which increases the potency and selectivity of the IL-2 agent for regulatory T cells (Treg) compared to other T cell types; (ii) is significantly stable, e.g., due to the presence of stabilizing V69A and Q74P mutations; (iii) has reduced or decreased binding capacity and/or binding affinity for CD25, which improves the lifetime of the IL-2 agent; (iv) does not substantially promote expansion, activation, survival, and/or proliferation of T effector cells and/or natural killer (NK) cells in vitro and/or in vivo', and/or (v) has reduced incorrect disulfide pairing and improved stability, e.g, due to the presence of the C125S mutation.
  • an IL-2 agent comprising the H16L mutation has reduced binding affinity for CD122 and/or CD132 and/or increased potency and selectivity for Treg over other T cell types, compared to an IL-2 agent comprising other H16 mutations.
  • an IL-2 fusion protein comprising amino acid substitutions H16L, V69A, Q74P, and C125S, has inter alia one or more (e.g., 2, 3, 4, 5, 6, 7, or all) of the following properties relative to a wild-type IL-2 or a reference IL-2 variant that does not comprise the amino acid substitutions: (i) enhanced or increased stability in vitro or in vivo', (ii) reduced or decreased binding capacity and/or binding affinity for human CD 122 in vitro and/or in vivo', (iii) reduced or decreased binding capacity and/or binding affinity for human CD 132 in vitro and/or in vivo', (iv) reduced or decreased affinity of the IL-2 variant for the heterodimeric IL-2 receptor composed of human CD122 and human CD132 (/.e.
  • human CD122/CD132 heterodimer in vitro and/or in vivo; (v) reduced or decreased or substantially unchanged binding capacity and/or binding affinity for human CD25 in vitro and/or in vivo; (vi) selective binding to regulatory T cells (e.g. Foxp3 + T cells); (vii) selective activation of the IL-2 signaling pathway in T regulatory cells (Tregs) in vitro or in vivo; or (viii) enhanced or increased ability to induce or promote Treg expansion, activity, survival and/or proliferation.
  • regulatory T cells e.g. Foxp3 + T cells
  • the IL-2 fusion protein comprises an IL-2 variant comprising an amino acid sequence chosen from: SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38,
  • the IL-2 fusion protein comprises an IL-2 variant comprising the amino acid sequence of SEQ ID NO: 4, or an amino acid sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 amino acids thereto.
  • the IL-2 fusion protein comprises an IL-2 variant comprising the amino acid sequence of SEQ ID NO: 5, or an amino acid sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 amino acids thereto.
  • the IL-2 fusion protein comprises the amino acid sequence of SEQ ID NO: 11, or an amino acid sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 amino acids thereto.
  • the IL-2 fusion protein comprises the amino acid sequence of SEQ ID NO: 1000, or an amino acid sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 amino acids thereto.
  • the IL-2 fusion protein comprises the amino acid sequence of SEQ ID NO: 1001, or an amino acid sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 amino acids thereto.
  • the IL-2 fusion protein comprises the amino acid sequence of SEQ ID NO: 1002, or an amino acid sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 amino acids thereto.
  • the IL-2 fusion protein comprises the amino acid sequence of any of SEQ ID NOs: 4, 5, 11, 1000, 1001, or 1002, or a functional fragment thereof. In an embodiment, the IL-2 fusion protein comprises the amino acid sequence of SEQ ID NO: 4 or 5, or a functional fragment thereof. In an embodiment, the IL-2 fusion protein comprises the amino acid sequence of SEQ ID NO: 4, or a functional fragment thereof. In an embodiment, the IL-2 fusion protein comprises the amino acid sequence of SEQ ID NO: 5, or a functional fragment thereof. In an embodiment, the IL-2 fusion protein comprises the amino acid sequence of SEQ ID NO: 11, or a functional fragment thereof.
  • the IL-2 fusion protein comprises the amino acid sequence of SEQ ID NO: 1000, or a functional fragment thereof. In an embodiment, the IL-2 fusion protein comprises the amino acid sequence of SEQ ID NO: 1001, or a functional fragment thereof. In an embodiment, the IL-2 fusion protein comprises the amino acid sequence of SEQ ID NO: 1002, or a functional fragment thereof.
  • an IL-2 fusion protein comprising the amino acid sequence of SEQ ID NO: 5, or a functional fragment thereof, can have at least one or more of the following advantageous properties: (i) has reduced binding affinity for CD122 and/or CD132, which increases the potency and selectivity of the IL-2 agent for regulatory T cells (Treg) compared to other T cell types; (ii) is significantly stable, e.g., due to the presence of stabilizing V69A and Q74P mutations; (iii) has reduced or decreased binding capacity and/or binding affinity for CD25, which improves the lifetime of the IL-2 agent; (iv) does not substantially promote expansion, activation, survival, and/or proliferation of T effector cells and/or natural killer (NK) cells in vitro and/or in vivo', and/or (v) has reduced incorrect disulfide pairing and improved stability, e.g., due to the presence of the C125S mutation.
  • an IL-2 agent comprising the H16L mutation has reduced binding affinity for CD 122 and/or CD 132 and/or increased potency and selectivity for Treg over other T cell types, compared to an IL-2 agent comprising other H16 mutations.
  • an IL-2 fusion protein comprising the amino acid sequence SEQ ID NO: 5, or a functional fragment thereof, or an amino acid sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 amino acids thereto, has inter alia one or more (e.g., 2, 3, 4, 5, 6, 7, or all) of the following properties relative to a wild-type IL-2 or a reference IL-2 fusion protein that does not comprise the amino acid substitutions: (i) enhanced or increased stability in vitro or in vivo', (ii) reduced or decreased binding capacity and/or binding affinity for human CD 122 in vitro and/or in vivo', (iii) reduced or decreased binding capacity and/or binding affinity for human CD 132 in vitro and/or in vivo; (iv) reduced or decreased affinity
  • human CD122/CD132 heterodimer in vitro and/or in vivo; (v) reduced or decreased or substantially unchanged binding capacity and/or binding affinity for human CD25 in vitro and/or in vivo; (vi) selective binding to regulatory T cells (e.g. Foxp3 + T cells); (vii) selective activation of the IL-2 signaling pathway in T regulatory cells (Tregs) in vitro or in vivo; or (viii) enhanced or increased ability to induce or promote Treg expansion, activity, survival and/or proliferation.
  • regulatory T cells e.g. Foxp3 + T cells
  • the IL-2 fusion proteins described herein comprise an Fc region, e.g. an Fc region having one or more mutations described herein, and/or having one or more structural or functional properties described herein.
  • Fc regions described herein can reduce (e.g., prevent) renal clearance and/or extend half-life of the IL-2 agents (e.g., via FcRn).
  • fusion protein refers to a protein, comprising two or more protein or peptide components.
  • the two or more protein or peptide components can be obtained from different sources or encoded by different genes.
  • a fusion protein is sometimes also referred to as a chimeric protein.
  • An Fc fusion protein (also known as Fc chimeric fusion protein, Fc-Ig, Ig-based chimeric fusion protein, or Fc-tag protein) can include an Fc region of an immunoglobulin (e.g., an Fc region described herein) linked (e.g., fused) to a protein or peptide.
  • the Fc region can be linked (e.g., fused genetically) to the protein or peptide directly, or indirectly, e.g., through a linker.
  • the Fc region is derived from the Fc region of IgG, e.g., human IgG, e.g., IgGl, IgG2, IgG3, or IgG4.
  • the Fc region is derived from the Fc region of IgGl, e.g., human IgGl.
  • An IL-2 fusion protein can include an IL-2 variant (e.g., an IL-2 variant described herein), or a functional fragment thereof, linked (e.g., fused) to a protein or peptide.
  • the IL-2 fusion protein is an IL-2-Fc fusion protein, e.g, further comprising an Fc region of an immunoglobulin (e.g., an Fc region described herein) linked (e.g., fused) to the IL-2 polypeptide (e.g., an IL-2 variant described herein) or a functional fragment thereof.
  • the IL-2 fusion protein is not an IL-2-Fc fusion protein, e.g., an IL-2 fusion variant described herein, or a functional fragment thereof, is linked (e.g., fused) to a protein or peptide other than an Fc region of IgG, e.g., human IgG, e.g., IgGl, IgG2, IgG3, or IgG4.
  • the IL-2 fusion protein comprises an amino acid sequence chosen from: SEQ ID NO: 56, SEQ ID NO: 57, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 61, SEQ ID NO: 62, SEQ ID NO: 63, SEQ ID NO: 64, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 67, SEQ ID NO: 68, SEQ ID NO: 69, SEQ ID NO: 70, SEQ ID NO: 71, SEQ ID NO: 72, SEQ ID NO: 73, SEQ ID NO: 74, SEQ ID NO: 75, SEQ ID NO: 76, SEQ ID NO: 77, SEQ ID NO: 78, SEQ ID NO: 79, SEQ ID NO: 80, SEQ ID NO: 81, SEQ ID NO: 82, SEQ ID NO: 83, SEQ ID NO: 84, SEQ ID NO: 85, SEQ ID NO:
  • the IL-2 fusion protein comprises an amino acid sequence chosen from: SEQ ID NO: 94, SEQ ID NO: 95, SEQ ID NO: 96, SEQ ID NO: 97, SEQ ID NO: 98, SEQ ID NO: 99, SEQ ID NO: 100, SEQ ID NO: 101, SEQ ID NO: 102, SEQ ID NO: 103, SEQ ID NO: 104, SEQ ID NO: 105, SEQ ID NO: 106, SEQ ID NO: 107, SEQ ID NO: 108, SEQ ID NO: 109, SEQ ID NO: 110, SEQ ID NO: 111, SEQ ID NO: 112, SEQ ID NO: 113, SEQ ID NO: 114, SEQ ID NO: 115, SEQ ID NO: 116, SEQ ID NO: 117, SEQ ID NO: 118, SEQ ID NO: 119, SEQ ID NO: 120, SEQ ID NO: 121, SEQ ID NO: 122, SEQ ID NO: 123, SEQ ID NO:
  • the IL-2 fusion protein comprises an amino acid sequence chosen from: SEQ ID NO: 132, SEQ ID NO: 133, SEQ ID NO: 134, SEQ ID NO: 135, SEQ ID NO: 136, SEQ ID NO: 137, SEQ ID NO: 138, SEQ ID NO: 139, SEQ ID NO: 140, SEQ ID NO: 141, SEQ ID NO: 142, SEQ ID NO: 143, SEQ ID NO: 144, SEQ ID NO: 145, SEQ ID NO: 146, SEQ ID NO: 147, SEQ ID NO: 148, SEQ ID NO: 149, SEQ ID NO: 150, SEQ ID NO: 151, SEQ ID NO: 152, SEQ ID NO: 153, SEQ ID NO: 154, SEQ ID NO: 155, SEQ ID NO: 156, SEQ ID NO: 157, SEQ ID NO: 158, SEQ ID NO: 159, SEQ ID NO: 160, SEQ ID NO: 161, SEQ ID NO
  • the IL-2 fusion protein comprises an amino acid sequence chosen from: SEQ ID NO: 170, SEQ ID NO: 171, SEQ ID NO: 172, SEQ ID NO: 173, SEQ ID NO: 174, SEQ ID NO: 175, SEQ ID NO: 176, SEQ ID NO: 177, SEQ ID NO: 178, SEQ ID NO: 179, SEQ ID NO: 180, SEQ ID NO: 181, SEQ ID NO: 182, SEQ ID NO: 183, SEQ ID NO: 184, SEQ ID NO: 185, SEQ ID NO: 186, SEQ ID NO: 187, SEQ ID NO: 188, SEQ ID NO: 189, SEQ ID NO: 190, SEQ ID NO: 191, SEQ ID NO: 192, SEQ ID NO: 193, SEQ ID NO: 194, SEQ ID NO: 195, SEQ ID NO: 196, SEQ ID NO: 197, SEQ ID NO: 198, SEQ ID NO: 199, SEQ ID NO:
  • the IL-2 fusion protein comprises an amino acid sequence chosen from: SEQ ID NO: 208, SEQ ID NO: 209, SEQ ID NO: 210, SEQ ID NO: 211, SEQ ID NO: 212, SEQ ID NO: 213, SEQ ID NO: 214, SEQ ID NO: 215, SEQ ID NO: 216, SEQ ID NO: 217, SEQ ID NO: 218, SEQ ID NO: 219, SEQ ID NO: 220, SEQ ID NO: 221, SEQ ID NO: 222, SEQ ID NO: 223, SEQ ID NO: 224, SEQ ID NO: 225, SEQ ID NO: 226, SEQ ID NO: 227, SEQ ID NO: 228, SEQ ID NO: 229, SEQ ID NO: 230, SEQ ID NO: 231, SEQ ID NO: 232, SEQ ID NO: 233, SEQ ID NO: 234, SEQ ID NO: 235, SEQ ID NO: 236, SEQ ID NO: 237, SEQ ID NO: 23
  • the IL-2 fusion protein comprises an amino acid sequence chosen from: SEQ ID NO: 246, SEQ ID NO: 247, SEQ ID NO: 248, SEQ ID NO: 249, SEQ ID NO: 250, SEQ ID NO: 251, SEQ ID NO: 252, SEQ ID NO: 253, SEQ ID NO: 254, SEQ ID NO: 255, SEQ ID NO: 256, SEQ ID NO: 257, SEQ ID NO: 258, SEQ ID NO: 259, SEQ ID NO: 260, SEQ ID NO: 261, SEQ ID NO: 262, SEQ ID NO: 263, SEQ ID NO: 264, SEQ ID NO: 265, SEQ ID NO: 266, SEQ ID NO: 267, SEQ ID NO: 268, SEQ ID NO: 269, SEQ ID NO: 270, SEQ ID NO: 271, SEQ ID NO: 272, SEQ ID NO: 273, SEQ ID NO: 274, SEQ ID NO: 275, SEQ ID NO: 276, SEQ ID NO: 277
  • the IL-2 fusion protein comprises an amino acid sequence chosen from: SEQ ID NO: 284, SEQ ID NO: 285, SEQ ID NO: 286, SEQ ID NO: 287, SEQ ID NO: 288, SEQ ID NO: 289, SEQ ID NO: 290, SEQ ID NO: 291, SEQ ID NO: 292, SEQ ID NO: 293, SEQ ID NO: 294, SEQ ID NO: 295, SEQ ID NO: 296, SEQ ID NO: 297, SEQ ID NO: 298, SEQ ID NO: 299, SEQ ID NO: 300, SEQ ID NO: 301, SEQ ID NO: 302, SEQ ID NO: 303, SEQ ID NO: 304, SEQ ID NO: 305, SEQ ID NO: 306, SEQ ID NO: 307, SEQ ID NO: 308, SEQ ID NO: 309, SEQ ID NO: 310, SEQ ID NO: 311, SEQ ID NO: 312, SEQ ID NO: 313, SEQ ID NO: 314, SEQ ID NO: 3
  • the IL-2 fusion protein comprises an amino acid sequence chosen from: SEQ ID NO: 322, SEQ ID NO: 323, SEQ ID NO: 324, SEQ ID NO: 325, SEQ ID NO: 326, SEQ ID NO: 327, SEQ ID NO: 328, SEQ ID NO: 329, SEQ ID NO: 330, SEQ ID NO: 331, SEQ ID NO: 332, SEQ ID NO: 333, SEQ ID NO: 334, SEQ ID NO: 335, SEQ ID NO: 336, SEQ ID NO: 337, SEQ ID NO: 338, SEQ ID NO: 339, SEQ ID NO: 340, SEQ ID NO: 341, SEQ ID NO: 342, SEQ ID NO: 343, SEQ ID NO: 344, SEQ ID NO: 345, SEQ ID NO: 346, SEQ ID NO: 347, SEQ ID NO: 348, SEQ ID NO: 349, SEQ ID NO: 350, SEQ ID NO: 351, SEQ ID NO: 351, S
  • the IL-2 fusion protein comprises an amino acid sequence chosen from: 1004, SEQ ID NO: 1005, SEQ ID NO: 1006, SEQ ID NO: 1007, SEQ ID NO: 1008, SEQ ID NO: 1009 or an amino acid sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 amino acids thereto.
  • the IL-2 fusion protein comprises the amino acid sequence of SEQ ID NO: 1004, or an amino acid sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 amino acids thereto.
  • the IL-2 fusion protein comprises the amino acid sequence of SEQ ID NO: 1005, or an amino acid sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 amino acids thereto.
  • the IL-2 fusion protein comprises the amino acid sequence of SEQ ID NO: 1006, or an amino acid sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 amino acids thereto.
  • the IL-2 fusion protein comprises the amino acid sequence of SEQ ID NO: 1007, or an amino acid sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 amino acids thereto.
  • the IL-2 fusion protein comprises the amino acid sequence of SEQ ID NO: 1008, or an amino acid sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 amino acids thereto.
  • the IL-2 agent comprises the amino acid sequence of any of SEQ ID NOs: 1004-1009, or a functional fragment thereof. In an embodiment, the IL-2 agent comprises the amino acid sequence of SEQ ID NO: 1007 or 1008, or a functional fragment thereof. In an embodiment, the IL-2 agent comprises the amino acid sequence of SEQ ID NO: 1004, or a functional fragment thereof. In an embodiment, the IL-2 agent comprises the amino acid sequence of SEQ ID NO: 1005, or a functional fragment thereof. In an embodiment, the IL-2 agent comprises the amino acid sequence of SEQ ID NO: 1006, or a functional fragment thereof. In an embodiment, the IL-2 agent comprises the amino acid sequence of SEQ ID NO: 1007, or a functional fragment thereof. In an embodiment, the IL-2 agent comprises the amino acid sequence of SEQ ID NO: 1008, or a functional fragment thereof. In an embodiment, the IL-2 agent comprises the amino acid sequence of SEQ ID NO: 1009, or a functional fragment thereof.
  • an IL-2 fusion protein comprising the amino acid sequence of SEQ ID NO: 1008, or a functional fragment thereof, can have at least one or more of the following advantageous properties: (i) has reduced binding affinity for CD 122 and/or CD 132, which increases the potency and selectivity of the IL-2 agent for regulatory T cells (Treg) compared to other T cell types; (ii) is significantly stable, e.g, due to the presence of stabilizing V69A and Q74P mutations; (iii) has reduced or decreased binding capacity and/or binding affinity for CD25, which improves the lifetime of the IL-2 agent; (iv) does not substantially promote expansion, activation, survival, and/or proliferation of T effector cells and/or natural killer (NK) cells in vitro and/or in vivo', (v) has reduced incorrect disulfide pairing and improved stability, e.g., due to the presence of the C125S mutation; and/or (vi) has reduced
  • an IL-2 agent comprising the H16L mutation has reduced binding affinity for CD 122 and/or CD 132 and/or increased potency and selectivity for Treg over other T cell types, compared to an IL-2 agent comprising other H16 mutations.
  • an IL-2 fusion protein comprising the amino acid sequence SEQ ID NO: 1008, or a functional fragment thereof, or an amino acid sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, or 50 amino acids thereto, has inter alia one or more (e.g., 2, 3, 4, 5, 6, 7, 8, or all) of the following properties relative to a wild-type IL-2 or a reference IL-2 fusion protein that does not comprise the amino acid substitutions: (i) enhanced or increased stability in vitro or in vivo', (ii) reduced or decreased binding capacity and/or binding affinity for human CD 122 in vitro and/or in vivo', (iii) reduced or decreased binding capacity and/or binding affinity for human CD 132 in vitro and/or in vivo
  • human CD122/CD132 heterodimer in vitro and/or in vivo', (v) reduced or decreased or substantially unchanged binding capacity and/or binding affinity for human CD25 in vitro and/or in vivo', (vi) selective binding to regulatory T cells (e.g. Foxp3 + T cells); (vii) selective activation of the IL-2 signaling pathway in T regulatory cells (Tregs) in vitro or in vivo', (viii) enhanced or increased ability to induce or promote Treg expansion, activity, survival and/or proliferation; or (ix) reduced or decreased effector function.
  • regulatory T cells e.g. Foxp3 + T cells
  • the IL-2 fusion protein comprises from N-terminus to C-terminus an IL-2 variant described herein and an Fc region (e.g., Fc region described herein).
  • the fusion protein further comprises a linker (e.g. , a linker described herein) between the IL-2 variant and the Fc region.
  • the IL-2 fusion forms a dimer, e.g., a homodimer.
  • the fusion protein comprises one or more glycosylation sites, or is glycosylated. In another embodiment, the fusion protein does not have a glycosylation site, or is not glycosylated.
  • the only amino acids in the fusion protein are canonical amino acids.
  • the fusion protein comprises naturally -occurring amino acids; analogs, derivatives and congeners thereof; amino acid analogs having variant side chains; and/or all stereoisomers of any of any of the foregoing.
  • the fusion protein may comprise the D- or L- optical isomers of amino acids and peptidomimetics.
  • this disclosure provides a method of making an IL-2 fusion protein disclosed herein.
  • the IL-2 fusion proteins described herein can be produced by any suitable recombinant DNA technique.
  • the method includes culturing a cell containing a nucleic acid encoding the IL-2 fusion protein under conditions that allow production of the fusion protein.
  • the method further includes isolating or purifying the IL-2 fusion protein.
  • the method further includes evaluating efficacy of the IL-2 fusion protein in a cell-based assay or in an animal model.
  • the method further includes administering the IL-2 fusion protein to a subject, e.g, a human.
  • nucleic acid molecule encoding an IL-2 fusion protein described herein, and vectors and host cells thereof.
  • the nucleic acid molecule includes, but is not limited to, RNA, genomic DNA and cDNA.
  • the IL-2 agent comprises an IL-2 complex, e.g., an IL-2 complex described herein.
  • the IL-2 complex is an IL-2/anti-IL-2 antibody immune complex (IL-2 ic).
  • IL-2 complexes such as IL-2/anti-IL-2 antibody immune complexes
  • the effect of IL-2 on cells e.g., Tregs
  • the mechanisms can include, e.g., the prolongation of the cytokine half-life in circulation.
  • IL-2 ic can selectively stimulate, for example, CD25high cells (e.g., IL-2/JES6-1 immune complexes), or CD122high cells (e.g., IL-2/S4B6 immune complexes).
  • CD25high cells e.g., IL-2/JES6-1 immune complexes
  • CD122high cells e.g., IL-2/S4B6 immune complexes
  • IL-2/JES6-1 immune complexes highly selectively stimulate regulatory T cells and they can be useful for transplantations and in treatment of autoimmune diseases.
  • IL-2/S4B6 immune complexes can have high stimulatory activity for NK cells and memory CD8+ T cells and they can replace the conventional IL-2 in cancer immunotherapy.
  • the IL-2 complex comprises an IL-2 variant described herein.
  • the IL-2 complex comprises one or more amino acid alterations (e.g., substitutions) described in Table 9.
  • the IL-2 complex comprises an amino acid sequence described in Table 9, or a functional fragment thereof.
  • the IL-2 complex comprises an anti-IL-2 antibody molecule.
  • the IL-2 complex comprises an IL-2 variant described herein and an anti-IL-2 antibody molecule.
  • the anti-IL-2 antibody molecule binds to the IL-2 variant.
  • the anti-IL-2 antibody molecule is capable of binding to the IL-2 variant and the wild-type IL-2.
  • the IL-2 variant comprises one or more mutations described herein. In an embodiment, the one or more mutations does not reduce, or does not substantially reduce, binding of the IL-2 variant to an anti-IL-2 antibody molecule.
  • the IL-2 complex comprises an amino acid sequence chosen from: SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 1000, SEQ ID NO: 1001,
  • the IL-2 complex modulates (e.g., stimulates) one or more activities of T cells.
  • the IL-2 complex stimulates CD25high cells.
  • the IL-2 complex stimulates Tregs.
  • the IL-2 complex stimulates CD122high cells.
  • the IL-2 complex stimulates NK cells and/or memory CD8+ T cells.
  • the IL-2 complex selectively stimulates CD25high cells over CD122high cells.
  • the IL-2 complex selectively stimulates CD122high cells over CD25high cells.
  • the IL-2 complex selectively stimulates Tregs over NK cells and/or memory CD8+ T cells.
  • the IL-2 complex selectively stimulates NK cells and/or memory CD8+ T cells over Tregs.
  • anti-IL-2 antibody molecules suitable for use are described, e.g., in International Application Publication No. WO 2016/164937, which is incorporated herein by reference in its entirety.
  • antibody molecule refers to a protein, e.g, an immunoglobulin chain or a fragment thereof, comprising at least one immunoglobulin variable domain sequence.
  • antibody molecule includes, for example, full-length, mature antibodies and antigen-binding fragments of an antibody.
  • an antibody molecule can include a heavy (H) chain variable domain sequence (abbreviated herein as VH), and a light (L) chain variable domain sequence (abbreviated herein as VL).
  • an antibody molecule in another example, includes two heavy (H) chain variable domain sequences and two light (L) chain variable domain sequence, thereby forming two antigen binding sites, such as Fab, Fab’, F(ab’)2, Fc, Fd, Fd’, Fv, single chain antibodies (scFv for example), single variable domain antibodies, diabodies (Dab) (bivalent and bispecific), and chimeric (e.g., humanized) antibodies, which may be produced by the modification of whole antibodies or those synthesized de novo using recombinant DNA technologies. These functional antibody fragments retain the ability to selectively bind with their respective antigen or receptor.
  • Antibodies and antibody fragments can be from any class of antibodies including, but not limited to, IgG, IgA, IgM, IgD, and IgE, and from any subclass (e.g., IgGl, IgG2, IgG3, and IgG4) of antibodies.
  • the antibody molecules can be monoclonal or polyclonal.
  • the antibody molecule can also be a human, humanized, CDR-grafted, or in vitro generated antibody.
  • the antibody molecule can have a heavy chain constant region chosen from, e.g, IgGl, IgG2, IgG3, or IgG4.
  • the antibody molecule can also have a light chain chosen from, e.g., kappa or lambda.
  • immunoglobulin (Ig) is used interchangeably with the term “antibody” herein.
  • antigen-binding fragments include: (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHI domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CHI domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a diabody (dAb) fragment, which consists of a VH domain; (vi) a camelid or camelized variable domain; (vii) a single chain Fv (scFv), see e.g., Bird et al.
  • antibody includes intact molecules as well as functional fragments thereof. Constant regions of the antibodies can be altered, e.g., mutated, to modify the properties of the antibody (e.g., to increase or decrease one or more of: Fc receptor binding, antibody glycosylation, the number of cysteine residues, effector cell function, or complement function).
  • the antibody molecule can be a single chain antibody.
  • a single-chain antibody (scFV) may be engineered (see, for example, Colcher, D. et al. (1999) Ann N Y Acad Sci 880:263-80; and Reiter, Y. (1996) Clin Cancer Res 2:245-52).
  • the single chain antibody can be dimerized or multimerized to generate multivalent antibodies having specificities for different epitopes of the same target protein.
  • the antibody molecules disclosed herein can also be single domain antibodies.
  • Single domain antibodies can include antibodies whose complementary determining regions are part of a single domain polypeptide. Examples include, but are not limited to, heavy chain antibodies, antibodies naturally devoid of light chains, single domain antibodies derived from conventional 4- chain antibodies, engineered antibodies and single domain scaffolds other than those derived from antibodies.
  • Single domain antibodies may be any of the art, or any future single domain antibodies.
  • Single domain antibodies may be derived from any species including, but not limited to mouse, human, camel, llama, fish, shark, goat, rabbit, and bovine.
  • a single domain antibody is a naturally occurring single domain antibody known as heavy chain antibody devoid of light chains. Such single domain antibodies are disclosed in WO 94/04678, for example.
  • variable domain derived from a heavy chain antibody naturally devoid of light chain is known herein as a VHH or nanobody to distinguish it from the conventional VH of four chain immunoglobulins.
  • VHH molecule can be derived from antibodies raised in Camelidae species, for example in camel, llama, dromedary, alpaca and guanaco. Other species besides Camelidae may produce heavy chain antibodies naturally devoid of light chain; such VHHs are also contemplated.
  • VH and VL regions can be subdivided into regions of hypervariability, termed “complementarity determining regions” (CDR), interspersed with regions that are more conserved, termed “framework regions” (FR or FW).
  • CDR complementarity determining region
  • FR framework regions
  • CDR complementarity determining region
  • each VH and VL typically includes three CDRs and four FRs, arranged from amino -terminus to carboxy -terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4.
  • an “immunoglobulin variable domain sequence” refers to an amino acid sequence which can form the structure of an immunoglobulin variable domain.
  • the sequence may include all or part of the amino acid sequence of a naturally -occurring variable domain.
  • the sequence may or may not include one, two, or more N- or C-terminal amino acids or may include other alterations that are compatible with formation of the protein structure.
  • antigen-binding region refers to the part of an antibody molecule that comprises determinants that form an interface that binds to an antigen, or an epitope thereof.
  • the antigen-binding region typically includes one or more loops (of at least, e.g, four amino acids or amino acid mimics) that form an interface that binds to the antigen.
  • the antigen-binding region of an antibody molecule includes at least one or two CDRs and/or hypervariable loops, or more typically at least three, four, five or six CDRs and/or hypervariable loops.
  • Compet or “cross-compete” are used interchangeably herein to refer to the ability of an antibody molecule to interfere with binding of another antibody molecule to a target.
  • the interference with binding can be direct or indirect (e.g, through an allosteric modulation of the antibody molecule or the target).
  • the extent to which an antibody molecule is able to interfere with the binding of another antibody molecule to the target, and therefore whether it can be said to compete, can be determined using a competition binding assay, for example, a FACS assay, an ELISA or BIACORE assay.
  • a competition binding assay is a quantitative competition assay.
  • a first antibody molecule is said to compete for binding to the target with a second antibody molecule when the binding of the first antibody molecule to the target is reduced by 10% or more, e.g., 20% or more, 30% or more, 40% or more, 50% or more, 55% or more, 60% or more, 65% or more, 70% or more, 75% or more, 80% or more, 85% or more, 90% or more, 95% or more, 98% or more, 99% or more in a competition binding assay (e.g., a competition assay described herein).
  • a competition binding assay e.g., a competition assay described herein.
  • monoclonal antibody or “monoclonal antibody composition” as used herein refer to a preparation of antibody molecules of single molecular composition.
  • a monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope.
  • a monoclonal antibody can be made by hybridoma technology or by methods that do not use hybridoma technology (e.g., recombinant methods).
  • An “effectively human” protein is a protein that does not evoke a neutralizing antibody response, e.g., the human anti-murine antibody (HAMA) response.
  • HAMA can be problematic in a number of circumstances, e.g., if the antibody molecule is administered repeatedly, e.g, in treatment of a chronic or recurrent disease condition.
  • a HAMA response can make repeated antibody administration potentially ineffective because of an increased antibody clearance from the serum (see, e.g., Saleh et al., Cancer Immunol. Immunother. 32:180-190 (1990)) and also because of potential allergic reactions (see, e.g., LoBuglio et al. , Hybridoma, 5:5117-5123 (1986)).
  • the antibody molecule can be a polyclonal or a monoclonal antibody.
  • the antibody can be recombinantly produced, e.g., produced by any suitable phage display or combinatorial methods.
  • the antibody molecule is a fully human antibody (e.g, an antibody made in a mouse which has been genetically engineered to produce an antibody from a human immunoglobulin sequence), or a non-human antibody, e.g, a rodent (mouse or rat), goat, primate (e.g, monkey), camel antibody.
  • the non-human antibody is a rodent (mouse or rat antibody). Methods of producing rodent antibodies are known in the art.
  • Human monoclonal antibodies can be generated using transgenic mice carrying the human immunoglobulin genes rather than the mouse system. Splenocytes from these transgenic mice immunized with the antigen of interest are used to produce hybridomas that secrete human mAbs with specific affinities for epitopes from a human protein (see e.g., Wood et al. International Application WO 91/00906, Kucherlapati et al. PCT publication WO 91/10741; Lonberg et al. International Application WO 92/03918; Kay et al. International Application 92/03917; Lonberg et al. 1994 Nature 368:856-859; Green, L.L. et al. 1994 Nature Genet.
  • An antibody can be one in which the variable region, or a portion thereof, e.g, the CDRs, are generated in a non-human organism, e.g., a rat or mouse. Chimeric, CDR-grafted, and humanized antibodies are within the invention. Antibodies generated in a non-human organism, e.g., a rat or mouse, and then modified, e.g, in the variable framework or constant region, to decrease antigenicity in a human are within the invention.
  • Chimeric antibodies can be produced by any suitable recombinant DNA technique.
  • Several are known in the art see Robinson et al., International Patent Publication PCT/US86/02269; Akira, et al., European Patent Application 184,187; Taniguchi, M., European Patent Application 171,496; Morrison et al., European Patent Application 173,494; Neuberger et al., International Application WO 86/01533; Cabilly et al. U.S. Patent No. 4,816,567; Cabilly et al., European Patent Application 125,023; Better et al. (1988 Science 240:1041-1043); Liu et al.
  • a humanized or CDR-grafted antibody will have at least one or two but generally all three recipient CDRs (of heavy and or light immunoglobulin chains) replaced with a donor CDR.
  • the antibody may be replaced with at least a portion of a non-human CDR or only some of the CDRs may be replaced with non-human CDRs. It is only necessary to replace the number of CDRs required for binding of the humanized antibody to lipopolysaccharide.
  • the donor will be a rodent antibody, e.g, a rat or mouse antibody
  • the recipient will be a human framework or a human consensus framework.
  • the immunoglobulin providing the CDRs is called the “donor” and the immunoglobulin providing the framework is called the “acceptor.”
  • the donor immunoglobulin is a non-human (e.g., rodent).
  • the acceptor framework is typically a naturally -occurring (e.g., a human) framework or a consensus framework, or a sequence about 85% or higher, e.g., 90%, 95%, 99% or higher identical thereto.
  • the term “consensus sequence” refers to the sequence formed from the most frequently occurring amino acids (or nucleotides) in a family of related sequences (See e.g, Winnaker, From Genes to Clones (Verlagsgesellschaft, Weinheim, Germany 1987). In a family of proteins, each position in the consensus sequence is occupied by the amino acid occurring most frequently at that position in the family. If two amino acids occur equally frequently, either can be included in the consensus sequence.
  • a “consensus framework” refers to the framework region in the consensus immunoglobulin sequence.
  • An antibody can be humanized by any suitable method, and several such methods known in the art (see e.g., Morrison, S. L., 1985, Science 229:1202-1207, by Oi et al., 1986, BioTechniques 4:214, and by Queen et al. US 5,585,089, US 5,693,761 and US 5,693,762, the contents of all of which are hereby incorporated by reference).
  • Humanized or CDR-grafted antibodies can be produced by CDR-grafting or CDR substitution, wherein one, two, or all CDRs of an immunoglobulin chain can be replaced. See e.g., U.S. Patent 5,225,539; Jones et al. 1986 Nature 321:552-525; Verhoeyan et al. 1988 Science 239:1534; Beidler et al. 1988 J. Immunol. 141:4053-4060; Winter US 5,225,539, the contents of all of which are hereby expressly incorporated by reference. Winter describes a CDR-grafting method which may be used to prepare humanized antibodies (UK Patent Application GB 2188638 A, filed on March 26, 1987; Winter US 5,225,539), the contents of which is expressly incorporated by reference.
  • humanized antibodies in which specific amino acids have been substituted, deleted or added. Criteria for selecting amino acids from the donor are described in, e.g., US 5,585,089, e.g., columns 12-16 of US 5,585,089, the contents of which are hereby incorporated by reference. Other techniques for humanizing antibodies are described in Padlan et al. EP 519596 Al, published on December 23, 1992.
  • the antibody molecule has a heavy chain constant region chosen from, e.g., the heavy chain constant regions of IgGl, IgG2 (e.g., IgG2a), IgG3, IgG4, IgM, IgAl, IgA2, IgD, and IgE; particularly, chosen from, e.g., the (e.g, human) heavy chain constant regions of IgGl, IgG2, IgG3, and IgG4.
  • the antibody molecule has a light chain constant region chosen from, e.g., the (e.g, human) light chain constant regions of kappa or lambda.
  • the constant region can be altered, e.g, mutated, to modify the properties of the antibody molecule (e.g., to increase or decrease one or more of: Fc receptor binding, antibody glycosylation, the number of cysteine residues, effector cell function, and/or complement function).
  • the antibody molecule has effector function and can fix complement.
  • the antibody molecule does not recruit effector cells or fix complement.
  • the antibody molecule has reduced or no ability to bind an Fc receptor.
  • it may be an isotype or subtype, fragment or other mutant, which does not support binding to an Fc receptor, e.g, it has a mutagenized or deleted Fc receptor binding region.
  • a constant region of the antibody molecule is altered.
  • Methods for altering an antibody constant region are known in the art.
  • Antibody molecules s with altered function e.g. altered affinity for an effector ligand, such as FcR on a cell, or the Cl component of complement can be produced by replacing at least one amino acid residue in the constant portion of the antibody with a different residue (see e.g., EP 388,151 Al, U.S. Pat. No. 5,624,821 and U.S. Pat. No. 5,648,260, the contents of all of which are hereby incorporated by reference).
  • Amino acid mutations which stabilize antibody structure such as S228P (EU nomenclature, S241P in Kabat nomenclature) in human IgG4 are also contemplated. Similar type of alterations could be described which if applied to the murine, or other species immunoglobulin would reduce or eliminate these functions.
  • the only amino acids in the antibody molecule are canonical amino acids.
  • the antibody molecule comprises naturally -occurring amino acids; analogs, derivatives and congeners thereof; amino acid analogs having variant side chains; and/or all stereoisomers of any of any of the foregoing.
  • the antibody molecule may comprise the D- or L- optical isomers of amino acids and peptidomimetics.
  • a polypeptide of an antibody molecule described herein may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non-amino acids.
  • the antibody molecule may also be modified; for example, by disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation, such as conjugation with a labeling component.
  • the polypeptide can be isolated from natural sources, can be a produced by recombinant techniques from a eukaryotic or prokaryotic host, or can be a product of synthetic procedures.
  • the antibody molecule described herein can be used alone in unconjugated form, or can be bound to a substance, e.g., a toxin or moiety (e.g., a therapeutic drug; a compound emitting radiation; molecules of plant, fungal, or bacterial origin; or a biological protein (e.g., a protein toxin) or particle (e.g., a recombinant viral particle, e.g., via a viral coat protein).
  • the antibody molecule can be coupled to a radioactive isotope such as an a-, P-, or y-emitter, or a P-and y-emitter.
  • an antibody molecule can be derivatized or linked to another functional molecule (e.g., another peptide or protein).
  • a “derivatized” antibody molecule is one that has been modified. Methods of derivatization include but are not limited to the addition of a fluorescent moiety, a radionucleotide, a toxin, an enzyme or an affinity ligand such as biotin. Accordingly, the antibody molecules are intended to include derivatized and otherwise modified forms of the antibodies described herein, including immunoadhesion molecules.
  • an antibody molecule can be functionally linked (by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other molecular entities, such as another antibody (e.g., a bispecific antibody or a diabody), a detectable agent, a toxin, a pharmaceutical agent, and/or a protein or peptide that can mediate association of the antibody or antibody portion with another molecule (such as a streptavidin core region or a polyhistidine tag).
  • another antibody e.g., a bispecific antibody or a diabody
  • detectable agent e.g., a toxin, a pharmaceutical agent, and/or a protein or peptide that can mediate association of the antibody or antibody portion with another molecule (such as a streptavidin core region or a polyhistidine tag).
  • Some types of derivatized antibody molecule are produced by crosslinking two or more antibodies (of the same type or of different types, e.g., to create bispecific antibodies).
  • Suitable crosslinkers include those that are heterobifunctional, having two distinctly reactive groups separated by an appropriate spacer (e.g., m-maleimidobenzoyl-N-hydroxysuccinimide ester) or homobifunctional (e.g., disuccinimidyl suberate).
  • Such linkers are available from Pierce Chemical Company, Rockford, Ill.
  • Exemplary fluorescent detectable agents include fluorescein, fluorescein isothiocyanate, rhodamine, 5dimethylamine-l-napthalenesulfonyl chloride, phycoerythrin and the like.
  • An antibody may also be derivatized with detectable enzymes, such as alkaline phosphatase, horseradish peroxidase, P -galactosidase, acetylcholinesterase, glucose oxidase and the like.
  • detectable enzymes such as alkaline phosphatase, horseradish peroxidase, P -galactosidase, acetylcholinesterase, glucose oxidase and the like.
  • detectable enzymes such as alkaline phosphatase, horseradish peroxidase, P -galactosidase, acetylcholinesterase, glucose oxidase and the like.
  • an antibody is derivatized with a detectable enzyme, it is detected by adding additional reagents that the enzyme uses to produce a detectable reaction product.
  • the detectable agent horseradish peroxidase is present, the addition of hydrogen peroxide and diaminobenzidine leads to a
  • an antibody may be derivatized with biotin, and detected through indirect measurement of avidin or streptavidin binding.
  • suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; and examples of bioluminescent materials include luciferase, luciferin, and aequorin.
  • Labeled antibody molecules can be used, for example, diagnostically and/or experimentally in a number of contexts, including (i) to isolate a predetermined antigen by standard techniques, such as affinity chromatography or immunoprecipitation; (ii) to detect a predetermined antigen (e.g., in a cellular lysate or cell supernatant) in order to evaluate the abundance and pattern of expression of the protein; (iii) to monitor protein levels in tissue as part of a clinical testing procedure, e.g, to determine the efficacy of a given treatment regimen.
  • a predetermined antigen e.g., in a cellular lysate or cell supernatant
  • An antibody molecule may be conjugated to another molecular entity, typically a label or a therapeutic (e.g., antimicrobial (e.g., antibacterial or bactericidal), immunomodulatory, immunostimularoty, cytotoxic, or cytostatic) agent or moiety.
  • Radioactive isotopes can be used in diagnostic or therapeutic applications. Radioactive isotopes that can be coupled to the antibody molecules include, but are not limited to a-, P-, or y-emitters, or P-and y-emitters.
  • radioactive isotopes include, but are not limited to iodine ( 131 I or 125 I), yttrium ( 90 Y), lutetium ( 177 Lu), actinium ( 225 Ac), praseodymium, astatine ( 211 At), rhenium ( 186 Re), bismuth ( 212 Bi or 213 Bi), indium ( in In), technetium (“mTc), phosphorus ( 32 P), rhodium ( 188 Rh), sulfur ( 35 S) , carbon ( 14 C), tritium ( 3 H), chromium ( 51 Cr), chlorine ( 36 C1), cobalt ( 57 Co or 58 Co), iron ( 59 Fe), selenium ( 75 Se), or gallium ( 67 Ga).
  • Radioisotopes useful as therapeutic agents include yttrium ( 90 Y), lutetium ( 177 Lu), actinium ( 225 Ac), praseodymium, astatine ( 211 At), rhenium ( 186 Re), bismuth ( 212 Bi or 213 Bi), and rhodium ( 188 Rh).
  • Radioisotopes useful as labels include iodine ( 131 I or 125 I), indium ( in In), technetium ( 99 mTc), phosphorus ( 32 P), carbon ( 14 C), and tritium ( 3 H), or one or more of the therapeutic isotopes listed above.
  • this disclosure provides a method of making an IL-2 complex described herein.
  • the method includes, e.g., contacting an IL-2 variant described herein with an anti-IL-2 antibody molecule (e.g., an anti-IL-2 antibody molecule that binds to the IL-2 variant), to thereby producing the IL-2 complex.
  • the method further comprises evaluating the efficacy of the IL-2 complex in vitro, ex vivo, or in vivo.
  • nucleic acid molecule encoding an IL-2 complex (or a portion thereol) described herein, and vectors and host cells thereof.
  • the nucleic acid molecule includes, but is not limited to, RNA, genomic DNA and cDNA.
  • the IL-2 agent comprises a conjugate, e.g., an IL-2 conjugate described herein.
  • the IL-2 conjugate comprises an IL-2 variant described herein and a non- IL-2 moiety.
  • the IL-2 conjugate comprises one or more amino acid alterations (e.g., substitutions) described in Table 9.
  • the IL-2 conjugate comprises an amino acid sequence described in Table 9, or a functional fragment thereof.
  • the non-IL-2 moiety comprises an antibody molecule, e.g., an antibody molecule described herein.
  • the non-IL-2 moiety comprises a polymer, e.g, a poly ether compound.
  • the polyether compound comprises polyethylene glycol (PEG).
  • the non-IL-2 moiety comprises a cytokine.
  • the IL-2 variant can be coupled to the non-IL-2 moiety directly, or indirectly, e.g, through a linker.
  • the IL-2 conjugate is an IL-2 fusion protein.
  • the IL-2 conjugate comprises an amino acid sequence chosen from: SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 1000, SEQ ID NO
  • the IL-2 conjugate is an immunoconjugate, e.g, comprising an antibody molecule.
  • the IL-2 variant is coupled to the antibody molecule by a covalent bond.
  • the IL-2 variant is coupled to the antibody molecule by a peptide bond.
  • the IL-2 variant and the antibody molecule forms a fusion protein.
  • the fusion protein comprises a linker between the IL-2 variant and the antibody molecule (e.g., a heavy chain, a light chain, or both).
  • the IL-2 variant is coupled to the antibody molecule by a non-peptide bond.
  • the IL-2 variant is not coupled to the antibody molecule by a non-peptide bond.
  • the IL-2 variant is coupled to the backbone of the antibody molecule. In another embodiment, the IL-2 variant is coupled to a side chain of the antibody molecule. In an embodiment, the antibody molecule is coupled to the backbone of the IL-2 variant. In an embodiment, the antibody molecule is coupled to a side-chain of the IL-2 variant.
  • two or more (e.g., three, four, five, six, seven, eight, or more) IL-2 variants are coupled to the antibody molecule.
  • four IL-2 variants are coupled to the antibody molecule.
  • the IL-2 variants can be the same, or at least some of the IL-2 variants are different from each other.
  • the IL-2 variant is coupled to the antibody molecule in a bivalent manner.
  • the IL-2 variant is coupled to the antibody molecule in a tetravalent manner.
  • the IL-2 conjugate is produced by enzymatic synthesis.
  • IL-2 conjugates can be produced by chemical synthesis of an IL-2 variant, expression of an antibody molecule, and enzymatic ligation of the IL-2 variant to the antibody molecule.
  • 90% or more, e.g., 92% or more, 95% or more, 97% or more, or 99% or more, reaction efficiency is achieved.
  • the method further comprises purifying the ADC.
  • the yield is 60% or more (e.g., 70% or more, 75% or more, 80% or more, 90% or more, or 95% or more) after purification.
  • the disclosure provides a combination of (a) an immunoconjugate comprising a first antibody molecule having a reduced effector function and an IL-2 variant described herein, and (b) a second antibody molecule having an increased effector function, for use in treating a disorder, e.g., a disorder described herein.
  • the reduced effector function of the first antibody comprises reduced binding to an activating Fc receptor, reduced ADCC, reduced ADCP, reduced CDC, reduced cytokine secretion, or a combination thereof.
  • the reduced effector function is reduced binding to an activating Fc receptor, e.g., a human Fc receptor.
  • the activating Fc receptor is an Fey receptor.
  • the activating Fc receptor is FcyRIIIa, FcyRI, or FcyRIIa.
  • the reduced effector function comprises reduced ADCC.
  • the increased effector function comprises reduced binding to an activating Fc receptor and reduced ADCC.
  • the first antibody molecule comprises one or more amino acid mutations (e.g., substitutions) in the Fc region as described herein.
  • the first antibody molecule comprises an amino acid substitution at position P329 of an immunoglobulin heavy chain.
  • the amino acid substitution comprises P329A or P329G, e.g., P329G.
  • the antibody molecule comprises a further amino acid substitution at a position of S228, E233, L234, L235, N297, P331, or a combination thereof, of an immunoglobulin heavy chain.
  • the further amino acid substitution comprises S228P, E233P, L234A, L235A, L235E, N297A, N297D, P331S, or a combination thereof.
  • the antibody comprises amino acid substitutions at positions P329, L234 and L235 of an immunoglobulin heavy chain.
  • the amino acid substitutions comprise L234A, L235A and P329G (LALA P329G).
  • the first antibody molecule is directed to an antigen presented on a tumor cell or in a tumor cell environment.
  • the first antibody is directed to an antigen chosen from Fibroblast Activation Protein (FAP), the Al domain of Tenascin-C (TNC Al), the A2 domain of Tenascin-C (TNC A2), the Extra Domain B of Fibronectin (EDB), Carcinoembryonic Antigen (CEA), and Melanoma-associated Chondroitin Sulfate Proteoglycan (MCSP).
  • the increased effector function of the second antibody molecule comprises increased binding to an activating Fc receptor, increased ADCC, increased ADCP, increased CDC, increased cytokine secretion, or a combination thereof.
  • the increased effector function comprises increased binding to an activating Fc receptor.
  • the activating Fc receptor is FcyRIIIa, FcyRI, or FcyRIIa.
  • the increased effector function comprises increased ADCC.
  • the increased effector function comprises increased binding to an activating Fc receptor and increased ADCC.
  • the second antibody molecule comprises one or more amino acid mutations (e.g., substitutions) in the Fc region.
  • the second antibody molecule comprises a modification of the glycosylation in the Fc region.
  • the modification of the glycosylation in the Fc region comprises an increased proportion of non-fucosylated oligosaccharides in the Fc region (e.g., increased to at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%) as compared to a non-modified antibody molecule.
  • the modification comprises an increased proportion of bisected oligosaccharides in the Fc region (e.g., increased to at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%), as compared to a non-modified antibody molecule.
  • the modification of the glycosylation in the Fc region comprises an increased proportion of bisected, non-fucosylated oligosaccharides in the Fc region (e.g., increased to at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%), as compared to a non-modified antibody molecule.
  • the second antibody molecule is directed to an antigen presented on a tumor cell.
  • the second antibody molecule is directed to an antigen chosen from CD20, Epidermal Growth Factor Receptor (EGFR), HER2, HER3, Insulin- like Growth Factor 1 Receptor (IGF-1R), c-Met, CUB domain-containing protein- 1 (CDCP1), Carcinoembryonic Antigen (CEA) and Melanoma-associated Chondroitin Sulfate Proteoglycan (MCSP).
  • the disease is a disorder treatable by stimulation of effector cell function.
  • the disclosure provides a composition comprising: (a) an immunoconjugate comprising a first antibody molecule having a reduced effector function and an IL-2 variant described herein, (b) a second antibody molecule having an increased effector function, and (c) a pharmaceutically acceptable carrier.
  • IL-2 agents e.g., IL-2 variants, IL-2 fusion proteins, IL-2 complexes, or IL-2 conjugates
  • IL-2R IL-2 receptor
  • IL-2R is a heterotrimeric protein expressed on the surface of certain immune cells, such as lymphocytes, that binds and responds to IL-2.
  • IL-2 receptor typically has three forms, generated by different combinations of three different chains: a (alpha) (also known as IL-2Ra, CD25, or Tac antigen), (beta) (also known as IL-2R , or CD122), and y (gamma) (also known as IL-2Ry, yc, common gamma chain, or CD 132).
  • the IL-2R chains are expressed separately and differently on various cell types and can assemble in different combinations and orders to generate low, intermediate, and high affinity IL-2Rs.
  • IL-2Ra binds IL-2 with low affinity;
  • IL-2R and IL-2Ry together form a complex that binds IL-2 with intermediate affinity (e.g., on memory T cells and NK cells);
  • IL-2Ra, IL-2R , and IL-2Ry together form a complex that binds IL-2 with high affinity (e.g., on activated T cells and regulatory T cells).
  • the binding of IL-2 to IL-2R can activate JAK1/JAK2 and initiate downstream intracellular signaling, e.g., the MAP kinase pathway, the Phosphoinositide 3-kinase (PI3K) pathway, or the JAK-STAT pathway (Liao et al., Curr Opin Immunol. 2011; 23(5): 598-604; Malek and Castro. Immunity. 2010; 33(2): 153-165).
  • IL-2R plays important roles in the immune system, tolerance and immunity. For example, the interaction between IL-2 and IL-2R is involved in promoting the differentiation of certain immature T cells into regulatory T cells, and the differentiation of T cells into effector T cells and into memory T cells. The interaction between IL-2 and IL-2R is also associated with autoimmune diseases, infections, and cell-mediated immunity.
  • the disclosure provides IL-2 agents comprising an IL-2 variant described herein that has an altered binding affinity to an IL-2R, e.g, one, two, or all of IL-2Ra, IL-2R , or IL-2Ry.
  • the IL-2 variant can have one or more (e.g., two, three, four, five, or more) amino acid alternations (e.g., substitutions or mutations) associated with the interaction between IL-2 and IL-2R, e.g., one, two, or all of IL-2Ra, IL-2R , or IL-2Ry.
  • the IL-2 agent has an altered (e.g., reduced) binding affinity to IL-2Ra.
  • the binding affinity to IL-2Ra is reduced by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more, relative to an IL-2 agent comprising a wild-type IL-2 or an IL-2 agent comprising a reference IL-2 variant.
  • the IL-2 agent has an altered (e.g., reduced) binding affinity to IL-2Rp.
  • the binding affinity to IL-2RP is reduced by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more, relative to an IL-2 agent comprising a wild-type IL-2 or an IL-2 agent comprising a reference IL-2 variant.
  • the IL-2 agent has an altered (e.g., reduced) binding affinity to IL-2Ry.
  • the binding affinity to IL-2Ry is reduced by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more, relative to an IL-2 agent comprising a wild-type IL-2 or an IL-2 agent comprising a reference IL-2 variant.
  • the IL-2 agent has an altered (e.g., reduced) binding affinity to IL-2Ra and an altered (e.g., reduced) binding affinity to IL-2Rp.
  • the binding affinity to IL- 2Ra is reduced by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more
  • the binding affinity to IL-2RP is reduced by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more.
  • the binding affinities to IL-2Ra and IL-2RP are reduced by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more, relative to an IL-2 agent comprising a wild-type IL-2 or an IL-2 agent comprising a reference IL-2 variant.
  • the IL-2 agent has an altered (e.g., reduced) binding affinity to IL-2Ra and an altered (e.g., reduced) binding affinity to IL-2Ry.
  • the binding affinity to IL- 2Ra is reduced by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more
  • the binding affinity to IL-2Ry is reduced by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more.
  • the binding affinities to IL-2Ra and IL-2Ry are reduced by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more, relative to an IL-2 agent comprising a wild-type IL-2 or an IL-2 agent comprising a reference IL-2 variant.
  • the IL-2 agent has an altered (e.g., reduced) binding affinity to IL-2RP and an altered (e.g., reduced) binding affinity to IL-2Ry.
  • the binding affinity to IL- 2RP is reduced by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more
  • the binding affinity to IL-2Ry is reduced by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more.
  • the binding affinities to IL-2RP and IL-2Ry are reduced by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more, relative to an IL-2 agent comprising a wild-type IL-2 or an IL-2 agent comprising a reference IL-2 variant.
  • the IL-2 agent has an altered (e.g., reduced) binding affinity to IL-2Ra, an altered (e.g., reduced) binding affinity to IL-2RP, and an altered (e.g., reduced) binding affinity to IL- 2Ry.
  • the binding affinity to IL-2Ra is reduced by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more
  • the binding affinity to IL-2RP is reduced by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more
  • the binding affinity to IL-2Ry is reduced by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more.
  • the binding affinities to IL-2Ra, IL-2RP, and IL-2Ry are reduced by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more, relative to an IL-2 agent comprising a wild-type IL-2 or an IL-2 agent comprising a reference IL-2 variant.
  • the binding affinity of an IL-2 agent provided by the disclosure to any of IL-2Ra, IL-2RP, or IL-2Ry is reduced, but not abolished.
  • the reduction can range from about 10% to about 90%, e.g., from about 20% to about 80%, from about 30% to about 70%, from about 40% to about 60%, from about 10% to about 50%, or from about 50% to about 90%, relative to an IL-2 agent comprising a wild-type IL-2 or an IL-2 agent comprising a reference IL-2 variant.
  • IL-2 agents e.g., IL-2 variants, fusion polypeptides, complexes, or immunoconjugates
  • an Fc region or a fragment thereof e.g., an Fc region, or a fragment thereof (e.g., a functional fragment thereol), described herein.
  • the IL-2 agent comprises an IL-2 variant described herein and an Fc region described herein. In an embodiment, the IL-2 agent further comprises a linker between the IL- 2 variant and the Fc region. In an embodiment, the IL-2 agent comprises an IL-2 fusion protein comprising an Fc region described herein. In an embodiment, the Fc region comprises one or more mutations described herein.
  • a fragment crystallizable region, or Fc region refers to a region of an immunoglobulin that interacts with an Fc receptor.
  • the Fc region interacts with a protein of the complement system. While without wishing to be bound by theory, it is believed that in an embodiment, the interaction between the Fc region with an Fc receptor, allows for activation of the immune system.
  • the naturally -occurring Fc region generally comprises two identical protein fragments, derived from the second and third constant domains of the antibody’s two heavy chains.
  • Naturally-occurring IgM and IgE Fc regions generally comprise three heavy chain constant domains (CH domains 2 ⁇ 1) in each polypeptide chain.
  • the Fc regions of IgGs can contain a highly conserved N-glycosylation site (Stadlmann et al. (2008). Proteomics 8 (14): 2858-2871; Stadlmann (2009) Proteomics 9 (17): 4143 4153).
  • glycosylation of the Fc fragment contributes to Fc receptor-mediated activities (Peipp et al. (2008) Blood 112 (6): 2390-2399).
  • the N-glycans attached to this site are predominantly core-fucosylated diantennary structures of the complex type.
  • small amounts of these N-glycans also contain bisecting GlcNAc and/or a-2,6 linked sialic acid residues.
  • SEQ ID NO: 40 An exemplary fragment of an Fc region amino acid sequence from human IgGl is provided in SEQ ID NO: 40 and is shown below: DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVWDVSHEDPEVKFNWYVDGVEVHNAK TKPREEQYGSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRD ELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFS CSVMHEALHNHYTQKSLSPGK ( SEQ ID NO: 40 ) In SEQ ID NO: 40, the first amino acid residue in this sequence is referred to as position 221 herein. The three histidine residues shown in bold and underlined are positions 310, 433 and 435, respectively.
  • An IL-2 agent comprising an Fc region or fragment thereof e.g., IL-2-Fc fusion protein
  • the Fc region comprises FcMutOOl. In an embodiment, the Fc region comprises FcMut002. In an embodiment, the Fc region comprises FcMut003. In an embodiment, the Fc region comprises FcMut004. In an embodiment, the Fc region comprises FcMut005. In an embodiment, the Fc region comprises FcMut006. In an embodiment, the Fc region comprises FcMut007. In an embodiment, the Fc region comprises FcMut008. In an embodiment, the Fc region comprises FcMut009. In an embodiment, the Fc region comprises FcMutOlO. In an embodiment, the Fc region comprises FcMutOl 1.
  • the Fc region comprises FcMutO12. In an embodiment, the Fc region comprises FcMutO13. In an embodiment, the Fc region comprises FcMutO14. In an embodiment, the Fc region comprises FcMut015. In an embodiment, the Fc region comprises FcMutO16. In an embodiment, the Fc region comprises FcMutO17. In an embodiment, the Fc region comprises FcMutO18. In an embodiment, the Fc region comprises FcMutO19. In an embodiment, the Fc region comprises FcMut020. In an embodiment, the Fc region comprises FcMutO21. In an embodiment, the Fc region comprises FcMutO22.
  • the Fc region comprises FcMutO23. In an embodiment, the Fc region comprises FcMutO24. In an embodiment, the Fc region comprises FcMutO26. In an embodiment, the Fc region comprises FcMutO27. In an embodiment, the Fc region comprises FcMutO28. In an embodiment, the Fc region comprises FcMutO29. In an embodiment, the Fc region comprises FcMut030. In an embodiment, the Fc region comprises FcMutO31. In an embodiment, the Fc region comprises FcMutO32. In an embodiment, the Fc region comprises FcMutO33. In an embodiment, the Fc region comprises FcMutO34.
  • the Fc region comprises FcMutO35. In an embodiment, the Fc region comprises FcMutO36. In an embodiment, the Fc region comprises FcMutO37. In an embodiment, the Fc region comprises FcMutO38. In an embodiment, the Fc region comprises FcMutO39. In an embodiment, the Fc region comprises FcMut040. In an embodiment, the Fc region comprises FcMutO41. In an embodiment, the Fc region comprises FcMutO42. In an embodiment, the Fc region comprises FcMutO43. In an embodiment, the Fc region comprises FcMutO44. In an embodiment, the Fc region comprises FcMutO45.
  • the Fc region comprises FcMutO46. In an embodiment, the Fc region comprises FcMutO47. In an embodiment, the Fc region comprises FcMutO48. In an embodiment, the Fc region comprises FcMutO49. In an embodiment, the Fc region comprises FcMut050. In an embodiment, the Fc region comprises FcMut051. In an embodiment, the Fc region comprises FcMutO52. In an embodiment, the Fc region comprises FcMutO53. In an embodiment, the Fc region comprises FcMutO67. In an embodiment, the Fc region comprises FcMutO68. In an embodiment, the Fc region comprises FcMutO69.
  • the Fc region comprises FcMut070. In an embodiment, the Fc region comprises FcMutO71. In an embodiment, the Fc region comprises FcMutO72. In an embodiment, the Fc region comprises FcMutO73. In an embodiment, the Fc region comprises FcMutO74. In an embodiment, the Fc region comprises FcMutO75. In an embodiment, the Fc region comprises FcMutO76. In an embodiment, the Fc region comprises FcMutO77. In an embodiment, the Fc region comprises FcMutO78. In an embodiment, the Fc region comprises FcMutO79. In an embodiment, the Fc region comprises FcMut080.
  • the Fc region comprises FcMutO81. In an embodiment, the Fc region comprises FcMutO82. In an embodiment, the Fc region comprises FcMutO83. In an embodiment, the Fc region comprises FcMutO84. In an embodiment, the Fc region comprises FcMutO85. In an embodiment, the Fc region comprises FcMutO86. In an embodiment, the Fc region comprises FcMutO87. In an embodiment, the Fc region comprises FcMutO88. In an embodiment, the Fc region comprises FcMutO89. In an embodiment, the Fc region comprises FcMut090. In an embodiment, the Fc region comprises FcMutO91.
  • the Fc region comprises FcMutO93. In an embodiment, the Fc region comprises FcMutO94. In an embodiment, the Fc region comprises FcMutO95. In an embodiment, the Fc region comprises FcMutO96. In an embodiment, the Fc region comprises FcMutO97. In an embodiment, the Fc region comprises FcMutO98. In an embodiment, the Fc region comprises FcMutO99. In an embodiment, the Fc region comprises FcMutlOO. In an embodiment, the Fc region comprises FcMutlOl. In an embodiment, the Fc region comprises FcMutlO2. In an embodiment, the Fc region comprises FcMutlO3.
  • the Fc region comprises FcMutlO4. In an embodiment, the Fc region comprises FcMutl05. In an embodiment, the Fc region comprises FcMutlO6. In an embodiment, the Fc region comprises FcMutlO7. In an embodiment, the Fc region comprises FcMutlO8. In an embodiment, the Fc region comprises FcMutlO9. In an embodiment, the Fc region comprises FcMutl 10. In an embodiment, the Fc region comprises FcMutl 11. In an embodiment, the Fc region comprises FcMutl 12. In an embodiment, the Fc region comprises FcMutl 13. In an embodiment, the Fc region comprises FcMutl 14.
  • the Fc region comprises FcMutl 15. In an embodiment, the Fc region comprises FcMutl 16. In an embodiment, the Fc region comprises FcMutl 17. In an embodiment, the Fc region comprises FcMutl 18. In an embodiment, the Fc region comprises FcMutl 19. In an embodiment, the Fc region comprises FcMutl20. In an embodiment, the Fc region comprises FcMutl21. In an embodiment, the Fc region comprises FcMutl22. In an embodiment, the Fc region comprises FcMutl23. In an embodiment, the Fc region comprises FcMutl24. In an embodiment, the Fc region comprises FcMutl25.
  • the Fc region comprises FcMutl26. In an embodiment, the Fc region comprises FcMutl27. In an embodiment, the Fc region comprises FcMutl28. In an embodiment, the Fc region comprises FcMutl29. In an embodiment, the Fc region comprises FcMutl30. In an embodiment, the Fc region comprises FcMutl31. In an embodiment, the Fc region comprises FcMutl32. In an embodiment, the Fc region comprises FcMutl33. In an embodiment, the Fc region comprises FcMutl34. In an embodiment, the Fc region comprises FcMutl35. In an embodiment, the Fc region comprises FcMutl36.
  • the Fc region comprises FcMutl37. In an embodiment, the Fc region comprises FcMutl38. In an embodiment, the Fc region comprises FcMutl39. In an embodiment, the Fc region comprises FcMutl40. In an embodiment, the Fc region comprises FcMutl41. In an embodiment, the Fc region comprises FcMutl42. In an embodiment, the Fc region comprises FcMutl43. In an embodiment, the Fc region comprises FcMutl44. In an embodiment, the Fc region comprises FcMutl45. In an embodiment, the Fc region comprises FcMutl46. In an embodiment, the Fc region comprises FcMutl47.
  • the Fc region comprises FcMutl48. In an embodiment, the Fc region comprises FcMutl49. In an embodiment, the Fc region comprises FcMutl50. In an embodiment, the Fc region comprises FcMutl51. In an embodiment, the Fc region comprises FcMutl52. In an embodiment, the Fc region comprises FcMutl53. In an embodiment, the Fc region comprises FcMutl 54. In an embodiment, the Fc region comprises FcMutl55. In an embodiment, the Fc region comprises FcMutl56. In an embodiment, the Fc region comprises FcMutl57. In an embodiment, the Fc region comprises FcMutl58.
  • the Fc region comprises FcMutl59. In an embodiment, the Fc region comprises FcMutl60. In an embodiment, the Fc region comprises FcMutl61. In an embodiment, the Fc region comprises FcMutl62. In an embodiment, the Fc region comprises FcMutl63. In an embodiment, the Fc region comprises FcMutl64. In an embodiment, the Fc region comprises FcMutl65. In an embodiment, the Fc region comprises FcMutl66. In an embodiment, the Fc region comprises FcMutl67. In an embodiment, the Fc region comprises FcMutl68. In an embodiment, the Fc region comprises FcMutl69.
  • the Fc region comprises FcMutl70. In an embodiment, the Fc region comprises FcMutl71. In an embodiment, the Fc region comprises FcMutl72. In an embodiment, the Fc region comprises FcMutl73. In an embodiment, the Fc region comprises FcMutl74. In an embodiment, the Fc region comprises FcMutl75. In an embodiment, the Fc region comprises FcMutl76. In an embodiment, the Fc region comprises FcMutl77. In an embodiment, the Fc region comprises FcMutl78. In an embodiment, the Fc region comprises FcMutl79. In an embodiment, the Fc region comprises FcMutl80.
  • the Fc region comprises FcMutl81. In an embodiment, the Fc region comprises FcMutl82. In an embodiment, the Fc region comprises FcMutl83. In an embodiment, the Fc region comprises FcMutl84. In an embodiment, the Fc region comprises FcMutl85. In an embodiment, the Fc region comprises FcMutl86. In an embodiment, the Fc region comprises FcMutl87. In an embodiment, the Fc region comprises FcMutl88. In an embodiment, the Fc region comprises FcMutl89. In an embodiment, the Fc region comprises FcMutl90. In an embodiment, the Fc region comprises FcMutl91.
  • the Fc region comprises FcMutl92. In an embodiment, the Fc region comprises FcMutl93. In an embodiment, the Fc region comprises FcMutl94. In an embodiment, the Fc region comprises FcMutl95. In an embodiment, the Fc region comprises FcMutl96. In an embodiment, the Fc region comprises FcMutl97. In an embodiment, the Fc region comprises FcMutl98. In an embodiment, the Fc region comprises FcMutl99. In an embodiment, the Fc region comprises FcMut200. In an embodiment, the Fc region comprises FcMut201. In an embodiment, the Fc region comprises FcMut202.
  • the Fc region comprises FcMut203. In an embodiment, the Fc region comprises FcMut204. In an embodiment, the Fc region comprises FcMut205. In an embodiment, the Fc region comprises FcMut206. In an embodiment, the Fc region comprises FcMut207. In an embodiment, the Fc region comprises FcMut208. In an embodiment, the Fc region comprises FcMut209. In an embodiment, the Fc region comprises FcMut210. In an embodiment, the Fc region comprises FcMut211. In an embodiment, the Fc region comprises FcMut212. In an embodiment, the Fc region comprises FcMut213. In an embodiment, the Fc region comprises FcMut214.
  • the Fc region comprises FcMut215. In an embodiment, the Fc region comprises FcMut216. In an embodiment, the Fc region comprises FcMut217. In an embodiment, the Fc region comprises FcMut218. In an embodiment, the Fc region comprises FcMut219. In an embodiment, the Fc region comprises FcMut220. In an embodiment, the Fc region comprises FcMut221. In an embodiment, the Fc region comprises FcMut222. In an embodiment, the Fc region comprises FcMut223. In an embodiment, the Fc region comprises FcMut224. In an embodiment, the Fc region comprises FcMut225.
  • the Fc region comprises FcMut226. In an embodiment, the Fc region comprises FcMut227. In an embodiment, the Fc region comprises FcMut228. In an embodiment, the Fc region comprises FcMut229. In an embodiment, the Fc region comprises FcMut230. In an embodiment, the Fc region comprises FcMut231. In an embodiment, the Fc region comprises FcMut232. In an embodiment, the Fc region comprises FcMut233. In an embodiment, the Fc region comprises FcMut234. In an embodiment, the Fc region comprises FcMut242. In an embodiment, the Fc region comprises FcMut243. In an embodiment, the Fc region comprises FcMut244.
  • the Fc region comprises one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, or more) of mutations or combinations of mutations chosen from FcMutO45, FcMutl71, FcMutl83, FcMutl86, FcMutl90, FcMutl97, FcMut213, FcMut215, FcMut216, FcMut219, FcMut222, FcMut223, FcMut224, FcMut226, FcMut227, FcMut228, or FcMut229.
  • FcMutO45 e.g., 2, 3, 4, 5, 6, 7, 8, 9, or more of mutations or combinations of mutations chosen from FcMutO45, FcMutl71, FcMutl83, FcMutl86, FcMutl90, FcMutl97, FcMut213,
  • the Fc region comprises one or more (e.g., 2, 3, 4, 5, 6, or all) of mutations or combinations of mutations chosen from FcMutO45, FcMutl83, FcMutl97, FcMut213, FcMut215, FcMut228, or FcMutl56.
  • the Fc region comprises one or more (e.g., 2, 3, 4, 5, or all) of mutations or combinations of mutations chosen from FcMutl83, FcMutl97, FcMut213, FcMut215, FcMut228, or FcMut229.
  • the Fc region does not comprise one or more (e.g., 2, 3, 4, or all) of mutations or combinations of mutations chosen from FcMutO18, FcMutO21, FcMut050, FcMutlO2, or YTE.
  • the Fc region comprises one or more (e.g., 2, 3, 4, or all) of mutations or combinations of mutations chosen from FcMutO18, FcMutO21, FcMut050, FcMutlO2, or YTE, and one or more other mutations or combinations of mutations described in Table 1.
  • the Fc region comprises one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more) of mutations or combinations of mutations described in Table 1 that result in a synergistic effect (e.g., binding affinity or circulating half-life) as described herein.
  • a synergistic effect e.g., binding affinity or circulating half-life
  • the Fc region comprises one or more (e.g., 2, 3, 4, 5, 6, or 7) mutations in residues chosen from T256, H285, N286, T307, Q311, N315, or A378. In an embodiment, the Fc region comprises one or more (e.g., 2, 3, 4, 5, 6, or 7) mutations chosen from T256D, H285N, N286D, T307Q, Q311V, N315D, or A378V.
  • the Fc region comprises a half-life enhancing mutation, a mutation that is capable of disrupting an Fc effector function, or both.
  • the Fc region comprises one or more mutations or combinations of mutations described herein, e.g., chosen from M252W, V308F/N434Y, R255Y, P257L/N434Y, V308F, P257N/M252Y, G385N, P257N/V308Y, N434Y, M252Y/S254T/T256E (“YTE”), M428L/N434S (“LS”), or any combination thereof.
  • the Fc region comprises (a) one or more (e.g., 2, 3, 4, 5, or all) combinations of mutations chosen from: T256D/Q311V/A378V, H285N/T307Q/N315D, H285D/T307Q/A378V, T307Q/Q311V/A378V, T256D/N286D/T307R/Q311V/A378V, or T256D/T307R/Q311V; (b) a mutation or a combination of mutations capable of disrupting an Fc effector function, e.g., N297G, L234A/L235A (also known as “LALA” mutation), L234A/L235A/P329G (also known as “LALAPG” mutation), or (c) both (a) and (b).
  • a mutation or a combination of mutations capable of disrupting an Fc effector function, e.g., N297G, L2
  • the Fc region comprises mutations T256D/Q311V/A378V and a mutation or a combination of mutations capable of disrupting an Fc effector function, e.g., L234A/L235A.
  • the Fc region comprises mutations H285N/T307Q/N315D and a mutation or a combination of mutations capable of disrupting an Fc effector function, e.g, L234A/L235A.
  • the Fc region comprises mutations H285D/T307Q/A378V and a mutation or a combination of mutations capable of disrupting an Fc effector function, e.g, L234A/L235A.
  • the Fc region comprises mutations T307Q/Q311 V/A378V and a mutation or a combination of mutations capable of disrupting an Fc effector function, e.g, L234A/L235A.
  • the Fc region comprises mutations T256D/N286D/T307R/Q311V/A378V and a mutation or a combination of mutations capable of disrupting an Fc effector function, e.g, L234A/L235A.
  • the Fc region comprises mutations T256D/T307R/Q311V and a mutation or a combination of mutations capable of disrupting an Fc effector function, e.g, L234A/L235A.
  • the Fc region comprises the Fc region of human IgGl, e.g, human IgGl m3 allotype. In an embodiment, the Fc region comprises the mutation N297G. In an embodiment, the Fc region comprises the Fc region of human IgGl allotype m3, human IgGl allotype m3 comprising the mutation N297G and/or other mutations of the Fc region of human IgGl allotype m3, or a fragment thereof.
  • the Fc region comprises the sequence of SEQ ID NO: 1003, or an amino acid sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 amino acids thereto.
  • any of the mutations in the Fc region that extend half-life described herein can be used in combination with any Fc mutation capable of enhancing or disrupting an Fc effector function.
  • the Fc region comprises the Fc region of human IgG4, human IgG4 containing S228P mutation, and/or R409K mutation, and/or other mutations of the Fc region of human IgG4, or a fragment thereof.
  • An exemplary fragment of an Fc region amino acid sequence from human IgG4 is provided in SEQ ID NO: 44 and is shown below: 3 O SIEKTI SKAK34OGQPREPQVYT35OLPPSQEEMTK36ONQVSLTCLVK37OGFYPSDIA 37 S VEWESNGQP
  • the first amino acid residue in this sequence is referred to as position 219 herein.
  • Mutations described to extend the half-life of human IgGl can be applied to human IgG4 Fc.
  • Mut215 corresponds to mutations T307Q/Q311V/A378V in SEQ ID NO: 44.
  • the Fc region can bind to various cell receptors (e.g., Fc receptors) and complement proteins.
  • the Fc region can also mediate different physiological effects of antibody molecules, e.g., detection of opsonized particles; cell lysis; degranulation of mast cells, basophils, and eosinophils; and other processes.
  • FcR Fc receptors
  • Fey receptors belong to the immunoglobulin superfamily, and are involved, e.g., in inducing phagocytosis of opsonized microbes.
  • This family includes several members, FcyR! (CD64), FcyRIIA (CD32), FcyRIIB (CD32), FcyRIIIA (CD 16a), FcyRIIIB (CD 16b), which differ in their antibody affinities due to their different molecular structure.
  • FcyRI can bind to IgG more strongly than FcyRII or FcyRIII does.
  • FcyRI also has an extracellular portion comprising three immunoglobulin (Ig)-like domains, one more domain than FcyRII or FcyRIII has. This property allows FcyRI to bind a sole IgG molecule (or monomer), but Fey receptors generally need to bind multiple IgG molecules within an immune complex to be activated.
  • Ig immunoglobulin
  • the Fey receptors differ in their affinity for IgG and the different IgG subclasses can have unique affinities for each of the Fey receptors. These interactions can be further tuned by the glycan (oligosaccharide) at certain position of IgG. For example, by creating steric hindrance, fucose containing CH2-84.4 glycans reduce IgG affinity for FcyRIIIA, whereas GO glycans, which lack galactose and terminate instead with GlcNAc moieties, have increased affinity for FcyRIIIA (Maverakis et al. (2015) Journal of Autoimmunity 57 (6): 1-13).
  • glycan oligosaccharide
  • the neonatal Fc receptor (FcRn) is expressed on multiple cell types and is similar in structure to MHC class I. This receptor also binds IgG and is involved in preservation of this antibody (Zhu et al. (2001). Journal of Immunology 166 (5): 3266-76.). FcRn is also involved in transferring IgG from a mother either via the placenta to her fetus or in milk to her suckling infant. This receptor may also play a role in the homeostasis of IgG serum levels.
  • FcaRI (or CD89) belongs to the FcaR subgroup.
  • FcaRI is found on the surface of neutrophils, eosinophils, monocytes, macrophages (including Kupffer cells), and dendritic cells. It comprises two extracellular Ig-like domains and is a member of both the immunoglobulin superfamily and the multi-chain immune recognition receptor (MIRR) family. It signals by associating with two FcRy signaling chains.
  • Fc-alpha/mu receptor (Fca/pR) is a type I transmembrane protein. It can bind IgA, although it has higher affinity for IgM (Shibuya and Hyundai (2006) Springer Seminars in Immunopathology 28 (4): 377-82). With one Ig-like domain in its extracellular portion, this Fc receptor is also a member of the immunoglobulin superfamily.
  • FceR There are two known types of FceR.
  • the high-affinity receptor FceRI is a member of the immunoglobulin superfamily (it has two Ig-like domains). FceRI is found on epidermal Langerhans cells, eosinophils, mast cells and basophils. This receptor can play a role in controlling allergic responses. FceRI is also expressed on antigen-presenting cells, and controls the production of immune mediators, e.g., cytokines that promote inflammation (von Bubnoff et al. (2003) Clinical and Experimental Dermatology 28 (2): 184-7).
  • the low-affinity receptor FceRII (CD23) is a C-type lectin. FceRII has multiple functions as a membrane-bound or soluble receptor. It can also control B cell growth and differentiation and blocks IgE-binding of eosinophils, monocytes, and basophils (Kikutani et al. (1989) Cib a Foundation Symposium 147: 23-31).
  • the Fc region can be engineered to contain an antigen-binding site to generate an Fcab fragment (Wozniak-Knopp et al. (2010) Protein Eng Des 23 (4): 289-297).
  • Fcab fragments can be inserted into a full immunoglobulin by swapping the Fc region, thus obtaining a bispecific antibody (with both Fab and Fcab regions containing distinct binding sites).
  • FcRn The binding and recycling of FcRn can be illustrated below.
  • IgG and albumin are internalized into vascular endothelial cells through pinocytosis.
  • the pH of the endosome is 6.0, facilitating association with membrane -bound FcRn.
  • the contents of endosomes can be processed in one of two ways: either recycling back to the apical cell membrane or transcytosis from the apical to the basolateral side. IgG not associated with FcRn is degraded by lysosomes.
  • FcRn interaction with IgG is mediated through Fc.
  • the binding of Fc to FcRn is pH specific, e.g, no significant binding at pH 7.4 and strong binding in acidic environment.
  • Structure of FcRn in complex with Fc domain of IgGl molecule is described, e.g, in FIG. 1 of International Application Publication No. WO2018/052556 or U.S. Application Publication No. US2018/0037634.
  • Each FcRn molecule generally binds to an Fc- monomer.
  • Fab domains can also influence binding of IgG to FcRn, e.g., have either a negative or no influence on the affinity of the IgG for FcRn.
  • FcRn binds proximal to the linker region between CH2 and CH3 domains of a Fc region. Modifications to the linker can impact Fc engagement with Fey receptors. Modifications on the Fc region can impact thermal stability and aggregation properties of the polypeptide.
  • compositions e.g., pharmaceutical compositions, which include an IL-2 agent described herein, and optionally a pharmaceutically acceptable carrier.
  • “pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • the carrier can be suitable for intravenous, intramuscular, subcutaneous, parenteral, rectal, spinal or epidermal administration (e.g., by injection or infusion).
  • less than about 5%, e.g., less than about 4%, 3%, 2%, or 1% of the IL-2 agents in the composition are present as aggregates.
  • At least about 95%, e.g, at least about 96%, 97%, 98%, 98.5%, 99%, 99.5%, 99.8%, or more of the IL-2 agents in the composition are present as monomers. In an embodiment, at least about 95%, e.g, at least about 96%, 97%, 98%, 98.5%, 99%, 99.5%, 99.8%, or more of the IL-2 agents in the composition are present as dimers. In an embodiment, the level of aggregates, dimers, or monomers is determined by chromatography, e.g., high performance liquid chromatography size exclusion chromatography (HPLC-SEC). In an embodiment, the IL-2 agent is formulated together with the pharmaceutically acceptable carrier.
  • HPLC-SEC high performance liquid chromatography size exclusion chromatography
  • compositions set out herein may be in a variety of forms. These include, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, liposomes, and suppositories.
  • a suitable form depends on the intended mode of administration and therapeutic application. Typical suitable compositions are in the form of injectable or infusible solutions.
  • One suitable mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal, intramuscular).
  • the IL-2 agent is administered by intravenous infusion or injection.
  • the IL-2 agent is administered by intramuscular or subcutaneous injection.
  • the IL-2 agent is administered subcutaneously (e.g., presented in an autoinjector or prefilled syringe).
  • parenteral administration and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrastemal injection and infusion.
  • compositions typically should be sterile and stable under the conditions of manufacture and storage.
  • the composition can be formulated as a solution, microemulsion, dispersion, liposome, or other ordered structure suitable to high antibody concentration.
  • Sterile injectable solutions can be prepared by incorporating the active compound (i.e., antibody or antibody portion) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-fdtered solution thereof.
  • the proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prolonged absorption of injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, monostearate salts and gelatin.
  • the IL-2 agents described herein can be administered by a variety of methods. Several are known in the art, and for many therapeutic, prophylactic, or diagnostic applications, an appropriate route/mode of administration is intravenous injection or infusion.
  • the IL-2 agents can be administered by intravenous infusion at a rate of less than lOmg/min; preferably less than or equal to 5 mg/min to reach a dose of about 1 to 100 mg/m 2 , preferably about 5 to 50 mg/m 2 , about 7 to 25 mg/m 2 and more preferably, about 10 mg/m 2 .
  • the route and/or mode of administration will vary depending upon the desired results.
  • the active compound may be prepared with a carrier that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems.
  • a controlled release formulation including implants, transdermal patches, and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, poly glycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker, Inc., New York, 1978.
  • the IL-2 agent is orally administered, for example, with an inert diluent or an assimilable edible carrier.
  • the IL-2 agent (and other ingredients, if desired) may also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated directly into the subject’s diet.
  • the IL-2 agent may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • To administer an IL-2 agent by other than parenteral administration it may be necessary to coat the compound with, or co-administer the compound with, a material to prevent its inactivation.
  • Therapeutic, prophylactic, or diagnostic compositions can also be administered with medical devices, and several are known in the art.
  • Dosage regimens are adjusted to provide the desired response (e.g., a therapeutic, prophylactic, or diagnostic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms is dictated by and directly dependent on (a) the unique characteristics of the molecule and the particular therapeutic, prophylactic, or diagnostic effect to be achieved, and (b) the limitations inherent in the art of compounding such a molecule for the treatment of sensitivity in individuals.
  • An exemplary, non-limiting range for a therapeutically, prophylactically, or diagnostically effective amount of an IL-2 agent is about 0.1-50 mg/kg, e.g, about 0.1-30 mg/kg, e.g., about 1-30, 1- 15, 1-10, 1-5, 5-10, or 1-3 mg/kg, e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, or 50 mg/kg.
  • the IL-2 agent can be administered by intravenous infusion at a rate of less than 10 mg/min, e.g., less than or equal to 5 mg/min to reach a dose of about 1 to 100 mg/m 2 , e.g., about 5 to 50 mg/m 2 , about 7 to 25 mg/m 2 , e.g., about 10 mg/m 2 . It is to be noted that dosage values may vary with the type and severity of the condition to be alleviated.
  • compositions herein may include a “therapeutically effective amount,” “prophylactically effective amount,” or “diagnostically effectively amount” of an IL-2 agent described herein.
  • a “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result.
  • a therapeutically effective amount of the polypeptide e.g., antibody molecule or fusion protein
  • a therapeutically effective amount may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the antibody or antibody portion to elicit a desired response in the individual.
  • a therapeutically effective amount is also one in which any toxic or detrimental effect of the antibody molecule is outweighed by the therapeutically beneficial effects.
  • a “therapeutically effective dosage” typically inhibits a measurable parameter by at least about 20%, e.g., by at least about 40%, by at least about 60%, or by at least about 80% relative to untreated subjects.
  • the measurable parameter may vary, e.g, based on the disordered being treated.
  • the ability of an IL-2 agent to inhibit a measurable parameter can be evaluated in an animal model system predictive of efficacy in treating or preventing a disorder described herein.
  • this property of a composition can be evaluated by examining the ability of the IL-2 agent to modulate a biological function of a target molecule or cell, e.g., by an in vitro assay.
  • a “prophylactically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.
  • a “diagnostically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired diagnostic result. Typically, a diagnostically effective amount is one in which a disorder, e.g., a disorder described herein, can be diagnosed in vitro, ex vivo, or in vivo.
  • the pharmaceutical composition is a good manufacturing practices (GMP)- grade pharmaceutical composition.
  • the pharmaceutical composition has greater than 99% purity, e.g., greater than 99.5%, 99.8%, or 99.9% purity. In an embodiment, greater than 50%, 60%, 70%, 80%, 90%, 95%, 98%, or 99% of the contaminants in the pharmaceutical composition are removed.
  • the pharmaceutical composition is in large scale, e.g. , at least 20g, 30g, 40g, 50g, 100g, 200g, 300g, 400g, 500g, 600g, 700g, 800g, 900g, 1000g, or more.
  • kits that comprise IL-2 agents described herein.
  • the kits can include one or more other elements including: instructions for use; other reagents, e.g., a label, a therapeutic agent, or an agent useful for chelating, or otherwise coupling, an antibody molecule coupled to a label or therapeutic agent, or a radioprotective composition; devices or other materials for preparing the IL-2 agent for administration; pharmaceutically acceptable carriers; and devices or other materials for administration to a subject.
  • the present disclosure also provides nucleic acids comprising a nucleotide sequence that encodes an IL-2 agent described herein.
  • the nucleic acid comprises a nucleotide sequence encoding an amino acid sequence of an IL-2 variant described herein, or a nucleotide sequence substantially identical thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein).
  • the nucleic acid comprises a nucleotide sequence encoding an IL-2 variant comprising one or more of the mutations described herein.
  • the nucleic acid further comprises a nucleotide sequence encoding an Fc region, e.g., an Fc region described herein, or having a nucleotide sequence substantially identical thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein).
  • the Fc region comprises one or more mutations, e.g, one or more mutations described herein.
  • the nucleic acid comprises from 5’ to 3’ a nucleotide sequence encoding an IL-2 variant described herein and a nucleotide sequence encoding an Fc region described herein.
  • the nucleic acid further comprises a nucleotide sequence encoding a linker, e.g, a linker described herein, or a nucleotide sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein).
  • the nucleic acid comprises from 5’ to 3’ a nucleotide sequence encoding an IL-2 variant described herein and a nucleotide sequence encoding a linker described herein.
  • the nucleic acid comprises from 5’ to 3’ a nucleotide sequence encoding a linker described herein, and a nucleotide sequence encoding an Fc region described herein.
  • the nucleic acid comprises a nucleotide sequence encoding an IL-2 fusion protein, e.g., an IL-2 fusion protein described herein, or a nucleotide sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein).
  • the nucleic acid encoding the IL-2 fusion protein comprises from 5’ to 3’ a nucleotide sequence encoding an IL-2 variant described herein and a nucleotide sequence encoding an Fc region described herein.
  • the nucleic acid encoding the IL-2 fusion protein comprises from 5’ to 3’ a nucleotide sequence encoding an IL-2 variant described herein, a nucleotide sequence encoding a linker described herein, and a nucleotide sequence encoding an Fc region described herein.
  • the nucleic acid comprises a portion of a nucleotide sequence described herein.
  • the portion may encode, for example, one, two, or all of an IL-2 variant, a linker, or an Fc region.
  • the nucleic acid comprises a nucleotide sequence encoding an amino acid sequence described in Table 9, or a functional fragment thereof. In an embodiment, the nucleic acid comprises a nucleotide sequence described in Table 10.
  • the nucleic acid comprises a nucleotide sequence encoding the amino acid sequence of any of SEQ ID NOs: 2-38 or 1000-1002, or a functional fragment thereof. In an embodiment, the nucleic acid comprises a nucleotide sequence encoding the amino acid sequence of any of SEQ ID NOs: 56-359 or 1004-1009, or a functional fragment thereof.
  • the nucleic acid comprises a nucleotide sequence of any of SEQ ID NOs: 361-398 or 1010-1012, or a nucleotide sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides thereto.
  • the nucleic acid further comprises a nucleotide sequence of any of SEQ ID NOs: 399-407 or 1013, or a nucleotide sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 nucleotides thereto.
  • the nucleic acid further comprises a nucleotide sequence of any of SEQ ID NOs: 408-415.
  • the nucleic acid comprises a nucleotide sequence of any of SEQ ID NOs: 416-481 or 1014-1019, or a nucleotide sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 nucleotides thereto.
  • the nucleic acid comprises a nucleotide sequence of any of SEQ ID NOs: 416-453 or 1014-1019.
  • the nucleic acid comprises a nucleotide sequence of any of SEQ ID NOs: 454-491. In an embodiment, the nucleic acid comprises a nucleotide sequence of any of SEQ ID NOs: 492-529. In an embodiment, the nucleic acid comprises a nucleotide sequence of any of SEQ ID NOs: 416-453. In an embodiment, the nucleic acid comprises a nucleotide sequence of any of SEQ ID NOs: 454-491. In an embodiment, the nucleic acid comprises a nucleotide sequence of any of SEQ ID NOs: 492-529. In an embodiment, the nucleic acid comprises a nucleotide sequence of any of SEQ ID NOs: 530-567.
  • the nucleic acid comprises a nucleotide sequence of any of SEQ ID NOs: 568-605. In an embodiment, the nucleic acid comprises a nucleotide sequence of any of SEQ ID NOs: 606-643. In an embodiment, the nucleic acid comprises a nucleotide sequence of any of SEQ ID NOs: 644-681.
  • the nucleic acid comprises the nucleotide sequence of any of SEQ ID NOs: 364, 365, 371, or 1010-1012, or a nucleotide sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides thereto.
  • the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 364.
  • the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 365.
  • the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 371. In an embodiment, the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 1010. In an embodiment, the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 1011. In an embodiment, the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 1012.
  • the nucleic acid further comprises the nucleotide sequence of SEQ ID NO: 1013, or a nucleotide sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, or 30 nucleotides thereto.
  • the nucleic acid further comprises the nucleotide sequence of SEQ ID NO: 48.
  • the nucleic acid comprises the nucleotide sequence of any of SEQ ID NOs: 1014-1017, or a nucleotide sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity thereof, or differing by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides thereto.
  • the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 1014.
  • the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 1015.
  • the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 1016. In an embodiment, the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 1017. In an embodiment, the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 1018. In an embodiment, the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 1019.
  • the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 364. In an embodiment, the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 365. In an embodiment, the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 371. In an embodiment, the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 1010. In an embodiment, the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 1011. In an embodiment, the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 1012. In an embodiment, the nucleic acid further comprises the nucleotide sequence of SEQ ID NO: 1013.
  • the nucleic acid further comprises the nucleotide sequence of SEQ ID NO: 48. In an embodiment, the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 1014. In an embodiment, the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 1015. In an embodiment, the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 1016. In an embodiment, the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 1017. In an embodiment, the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 1018. In an embodiment, the nucleic acid comprises the nucleotide sequence of SEQ ID NO: 1019.
  • the nucleic acids disclosed herein include deoxyribonucleotides or ribonucleotides, or analogs thereof.
  • the polynucleotide may be either single-stranded or double -stranded, and if singlestranded may be the coding strand or non-coding (antisense) strand.
  • a polynucleotide may comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs.
  • the sequence of nucleotides may be interrupted by non-nucleotide components.
  • a polynucleotide may be further modified after polymerization, such as by conjugation with a labeling component.
  • the nucleic acid may be a recombinant polynucleotide, or a polynucleotide of genomic, cDNA, semisynthetic, or synthetic origin which either does not occur in nature or is linked to another polynucleotide in a nonnatural arrangement.
  • the disclosure features host cells and vectors comprising the nucleic acids described herein.
  • the nucleic acids may be present in a single vector or separate vectors present in the same host cell or separate host cell, as described in more detail below.
  • the disclosure features methods of treating a disorder (e.g., a disorder described herein) comprising administering to a subject in need thereof an effective amount of a nucleic acid described herein.
  • a disorder e.g., a disorder described herein
  • administering to a subject in need thereof an effective amount of a nucleic acid described herein.
  • the present disclosure features vectors that comprise a nucleotide sequence encoding an IL-2 agent described herein.
  • the vector comprises a nucleic acid described herein e.g., in Table 10).
  • the vector comprises a nucleotide sequence encoding an amino acid sequence of an IL-2 variant described herein (e.g., in Table 9), or a nucleotide sequence substantially identical thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein).
  • the vector comprises a nucleotide sequence encoding an IL-2 variant comprising one or more of the mutations described herein.
  • the vector further comprises a nucleotide sequence encoding an Fc region, e.g., an Fc region described herein, or having a nucleotide sequence substantially identical thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein).
  • the Fc region comprises one or more mutations, e.g, one or more mutations described herein.
  • the vector comprises from 5’ to 3’ a nucleotide sequence encoding an IL-2 variant described herein and a nucleotide sequence encoding an Fc region described herein.
  • the vector further comprises a nucleotide sequence encoding a linker, e.g., a linker described herein, or a nucleotide sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein).
  • the vector comprises from 5’ to 3’ a nucleotide sequence encoding an IL-2 variant described herein and a nucleotide sequence encoding a linker described herein.
  • the vector comprises from 5 ’ to 3 ’ a nucleotide sequence encoding a linker described herein, and a nucleotide sequence encoding an Fc region described herein.
  • the vector comprises a nucleotide sequence encoding an IL-2 fusion protein, e.g., an IL-2 fusion protein described herein, or a nucleotide sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein).
  • the vector encoding the IL-2 fusion protein comprises from 5’ to 3’ a nucleotide sequence encoding an IL-2 variant described herein and a nucleotide sequence encoding an Fc region described herein.
  • the vector encoding the IL-2 fusion protein comprises from 5’ to 3’ a nucleotide sequence encoding an IL-2 variant described herein, a nucleotide sequence encoding a linker described herein, and a nucleotide sequence encoding an Fc region described herein.
  • the vector further comprises a nucleotide sequence encoding a heavy chain variable region of an anti-IL-2 antibody molecule, e.g., an anti-IL-2 antibody molecule described herein.
  • the vector further comprises a nucleotide sequence encoding a light chain variable region of an anti-IL-2 antibody molecule, e.g., an anti-IL-2 antibody molecule described herein.
  • the vector further comprises a nucleotide sequence encoding a heavy chain variable region and a light chain variable region of an anti-IL-2 antibody molecule, e.g, an anti-IL-2 antibody molecule described herein.
  • the vector further comprises a nucleotide sequence encoding at least one, two, or three CDRs from a heavy chain variable region of an anti-IL-2 antibody molecule, e.g., an anti-IL-2 antibody molecule described herein.
  • the vector further comprises a nucleotide sequence encoding at least one, two, or three CDRs from a light chain variable region of an anti-IL-2 antibody molecule, e.g., an anti-IL-2 antibody molecule described herein.
  • the vector comprises a nucleotide sequence encoding at least one, two, three, four, five, or six CDRs from heavy and light chain variable regions of an anti-IL-2 antibody molecule, e.g., an anti-IL-2 antibody molecule described herein.
  • the vector comprises a portion of a nucleotide sequence described herein.
  • the portion may encode, for example, an IL-2 variant; a liker an Fc region; a variable region (e.g., VH or VL); one, two, or three or more (e.g., four, five, or six) CDRs; or one, two, three, or four or more framework regions.
  • the vectors include, but are not limited to, a virus, plasmid, cosmid, lambda phage or a yeast artificial chromosome (Y AC).
  • vectors utilize DNA elements which are derived from animal viruses such as, for example, bovine papilloma virus, polyoma virus, adenovirus, vaccinia virus, baculovirus, retroviruses (Rous Sarcoma Virus, MMTV or MOMLV) or SV40 virus.
  • DNA elements which are derived from animal viruses such as, for example, bovine papilloma virus, polyoma virus, adenovirus, vaccinia virus, baculovirus, retroviruses (Rous Sarcoma Virus, MMTV or MOMLV) or SV40 virus.
  • RNA elements derived from RNA viruses such as Semliki Forest virus, Eastern Equine Encephalitis virus and Flaviviruses.
  • cells which have stably integrated the DNA into their chromosomes may be selected by introducing one or more markers which allow for the selection of transfected host cells.
  • the marker may provide, for example, prototropy to an auxotrophic host, biocide resistance (e.g, antibiotics), or resistance to heavy metals such as copper, or the like.
  • the selectable marker gene can be either directly linked to the DNA sequences to be expressed or introduced into the same cell by cotransformation. Additional elements may also be needed for optimal synthesis of mRNA. These elements may include splice signals, as well as transcriptional promoters, enhancers, and termination signals.
  • the expression vectors may be transfected or introduced into an appropriate host cell.
  • Various techniques may be employed to achieve this, such as, for example, protoplast fusion, calcium phosphate precipitation, electroporation, retroviral transduction, viral transfection, gene gun, lipid- based transfection or other conventional techniques.
  • protoplast fusion the cells are grown in media and screened for the appropriate activity.
  • Methods and conditions for culturing the resulting transfected cells and for recovering the polypeptides (e.g., IL-2 variants or IL-2 fusion proteins) produced are known to those skilled in the art and may be varied or optimized depending upon the specific expression vector and mammalian host cell employed, based upon the present description.
  • the present disclosure also provides cells comprising a nucleic acid or vector encoding an IL-
  • the cell is a host cell.
  • the host cell can comprise an IL-2 agent engineered in accordance with a method described herein.
  • the cell is an isolated cell.
  • the cell is a cultured cell.
  • the cell comprises a nucleic acid comprising a nucleotide sequence encoding an IL-2 agent described herein (e.g., in Table 10), a nucleotide sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein), or a portion of the aforesaid nucleic acid.
  • a nucleic acid comprising a nucleotide sequence encoding an IL-2 agent described herein (e.g., in Table 10), a nucleotide sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein), or a portion of the aforesaid nucleic acid.
  • the cell comprises a vector comprising a nucleotide sequence encoding an IL-2 agent described herein, a nucleotide sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein), or a portion of the aforesaid vector.
  • the cell is genetically engineered to comprise a nucleic acid or vector encoding an IL-2 agent described herein.
  • the host cells are genetically engineered by using an expression cassette.
  • expression cassette refers to nucleotide sequences, which are capable of affecting expression of a gene in hosts compatible with such sequences.
  • cassettes may include a promoter, an open reading frame with or without introns, and a termination signal. Additional factors necessary or helpful in effecting expression may also be used, for example, an inducible promoter.
  • the cell can be, but is not limited to, a eukaryotic cell, a bacterial cell, an insect cell, or a human cell.
  • Suitable eukaryotic cells include, but are not limited to, Vero cells, HeLa cells, COS cells, CHO cells, HEK293 cells, BHK cells and MDCKII cells.
  • Suitable insect cells include, but are not limited to, Sf9 cells.
  • IL-2 agents e.g., IL-2 variants, fusion polypeptides, complexes, or immunoconjugates
  • compositions described herein and the nucleic acids described herein have in vitro, ex vivo, and in vivo therapeutic, prophylactic, and/or diagnostic utilities.
  • the IL-2 agent modulates (e.g., reduces (e.g., inhibits, blocks, or neutralizes) or increases (e.g., activates, initiates, or enhances)) one or more biological activities associated with IL-2.
  • these IL-2 agents can be administered to cells in culture, in vitro or ex vivo, or to a subject, e.g., a human subject, e.g., in vivo, to modulate one or more biological activities associated with IL-2.
  • the disclosure provides a method of treating, preventing, or diagnosing a disorder, e.g., a disorder described herein, in a subject, comprising administering to the subject an IL-2 agent described herein, such that the disorder is treated, prevented, or diagnosed.
  • a method comprising contacting the IL-2 agent described herein with cells in culture, e.g., in vitro or ex vivo, or administering the IL-2 agent described herein to a subject, e.g., in vivo, to treat, prevent, or diagnose a disorder, e.g., a disorder associated with IL-2 (e.g., a disorder described herein).
  • the term “subject” is intended to include human and non-human animals.
  • the subject is a human subject, e.g., a human patient having a disorder described herein, or at risk of having a disorder described herein.
  • non-human animals includes mammals and non-mammals, such as non-human primates.
  • the subject is a human.
  • the methods and compositions described herein are suitable for treating human patients for a disorder described herein.
  • Patients having a disorder described herein include those who have developed a disorder described herein but are (at least temporarily) asymptomatic, patients who have exhibited a symptom of a disorder described herein, or patients having a disorder related to or associated with a disorder described herein.
  • the IL-2 agents described herein selectively stimulate regulatory T cells (Tregs).
  • the IL-2 agents described herein can promotes the proliferation, survival, activation, and/or function of CD3+FoxP3+ T cells over CD3+FoxP3- T cells.
  • Methods of measuring the ability to selectively stimulate Tregs can be measured by flow cytometry of peripheral blood leukocytes, in which there is an observed increase in the percentage of FOXP3+CD4+ T cells among total CD4+ T cells, an increase in percentage of FOXP3+CD8+ T cells among total CD8+ T cells, an increase in percentage of FOXP3+ T cells relative to NK cells, and/or a greater increase in the expression level of CD25 on the surface of FOXP3+ T cells relative to the increase of CD25 expression on other T cells.
  • Treg cells can also be detected as increased representation of demethylated FOXP3 promoter DNA (i.e., the Treg-specific demethylated region, or TSDR) relative to demethylated CD3 genes in DNA extracted from whole blood, as detected by sequencing of polymerase chain reaction (PCR) products from bisulfite -treated genomic DNA (J. Sehouli, et al. 2011. Epigenetics 6:2, 236-246).
  • PCR polymerase chain reaction
  • the IL-2 agents described agents can achieve immune modulation through selective activation of regulatory T cells, resulting in T reg stimulation with minimal effect on T effector and NK cells.
  • the IL-2 agents described herein are particularly suitable for treating transplant rejection, e.g., heart transplant rejection.
  • the IL-2 agent results in immune modulation without immunosuppression, which is highly desired in an IL-2 therapy.
  • the disclosure provides a method of increasing the ratio of regulatory T cells (Tregs) to non-regulatory T cells (non-Tregs) within a population of T cells, comprising contacting the population of T cells with an effective amount of an IL-2 agent described herein.
  • the IL-2 agent selectively increases the ratio of Tregs over non-Tregs by about 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more, or about 2, 3, 4, 5, 6, 7, 8, 9, 10- fold or more.
  • the IL-2 agent selectively increases the ratio of CD3+FoxP3+ cells to CD3+FoxP3- cells by about 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more, or about 2, 3, 4, 5, 6, 7, 8, 9, 10-fold or more.
  • the disclosure provides a method of increasing the ratio of regulatory T cells (Tregs) to non-regulatory T cells (non-Tregs) in a subject (e.g., in the peripheral blood of a subject), comprising contacting the subject or sample with an effective amount of an IL-2 agent described herein.
  • a subject e.g., in the peripheral blood of a subject
  • the IL-2 agent selectively increases the ratio of Tregs over non-Tregs in the subject, or in a sample (e.g., a peripheral blood sample) from the subject, by about 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more, or about 2, 3, 4, 5, 6, 7, 8, 9, 10-fold or more.
  • the IL-2 agent selectively increases the ratio of CD3+FoxP3+ cells to CD3+FoxP3- cells in the subject, or in a sample (e.g., a peripheral blood sample) from the subject, by about 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more, or about 2, 3, 4, 5, 6, 7, 8, 9, 10-fold or more.
  • the disclosure provides a method of increasing the ratio of regulatory T cells (Tregs) to natural killer cells (NKs) in a subject (e.g., in the peripheral blood of a subject), comprising contacting the subject or sample with an effective amount of an IL-2 agent described herein.
  • Tregs regulatory T cells
  • NKs natural killer cells
  • the IL-2 agent selectively increases the ratio of Tregs over NKs in the subject, or in a sample (e.g., a peripheral blood sample) from the subject, by about 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more, or about 2, 3, 4, 5, 6, 7, 8, 9, 10-fold or more.
  • the IL-2 agent selectively increases the ratio of CD3+FoxP3+ cells to CD3-CD19- lymphocytes expressing CD56 and/or CD16 in the subject, or in a sample (e.g., a peripheral blood sample) from the subject, by about 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more, or about 2, 3, 4, 5, 6, 7, 8, 9, 10-fold or more.
  • IL-2 agents e.g., IL-2 variants, fusion polypeptides, complexes, or immunoconjugates
  • pharmaceutical compositions disclosed herein and the nucleic acids described herein can be used to treat or prevent various disorders or conditions.
  • the disorder is a transplant rejection. In an embodiment, the disorder is a heart transplant rejection. In an embodiment, the disorder is a heart disease, e.g., a heart disease that can be treated by a heart transplant.
  • the IL-2 agents described herein can have an optimal or improved half-life, which can be desirable for treating or preventing a wide range of disorders or conditions. While not wishing to be bound by theory, it is believed that in an embodiment, the IL-2 agents described herein can provide one or more benefits over another IL-2 agent having the same or similar binding affinity and/or specificity (e.g., an IL-2 agent that does not have, or has not been engineered to have, an optimal or improved half-life). These benefits can include, but are not limited to, an increased therapeutic or preventive efficacy, a reduced dosage regimen, or an improved pharmacokinetic property. In an embodiment, the IL-2 includes a mutated Fc region as described herein.
  • the ratio of regulatory T cells (Tregs) to non-regulatory T cells within the subject increases after the administration.
  • the ratio of regulatory T cells (Tregs) to non-regulatory T cells within the subject (e.g., in the peripheral blood of the subject) remains essentially the same after the administration.
  • the method further comprises identifying a subject who needs an increased level of Tregs.
  • the method further comprises determining the level of Tregs in the subject prior to and/or after the administration.
  • the IL-2 agents described herein are typically administered at a frequency that keeps a therapeutically effective level of IL-2 agents in the patient’s system until the patient recovers.
  • the IL-2 agents may be administered at a frequency that achieves a serum concentration sufficient for at least about 1, 2, 5, 10, 20, 30, or 40 agents to bind each target molecule or cell.
  • the IL-2 agent is administered every 1, 2, 3, 4, 5, 6, or 7 days, every 1, 2, 3, 4, 5, or 6 weeks, or every 1, 2, 3, 4, 5, or 6 months.
  • the IL-2 agent is administered once a month.
  • the IL-2 agent is administered once a week.
  • agents e.g., antibody molecules or fusion proteins
  • Suitable dosages of the agents used will depend on the age and weight of the subject and the particular drug used.
  • the ratio of regulatory T cells (Tregs) to non-regulatory T cells within the subject increases after the administration. In an embodiment, the ratio of regulatory T cells (Tregs) to non-regulatory T cells within the subject (e.g., in the peripheral blood of the subject) remains essentially the same after the administration.
  • the IL-2 agents can be used by themselves or conjugated to a second agent, e.g., a protein, e.g., an antibody molecule, a polymer (e.g., polyethylene glycol (PEG)), or a cytokine.
  • a second agent e.g., a protein, e.g., an antibody molecule, a polymer (e.g., polyethylene glycol (PEG)), or a cytokine.
  • the second agent comprises a second IL-2 agent. This method includes administering the IL-2 agent, alone or conjugated to a second agent, to a subject requiring such treatment.
  • IL-2 agents e.g., e.g., IL-2 variants, IL-2 fusion proteins, IL-2 complexes, or IL-2 conjugates
  • IL-2 agents can be used in combination with other therapies.
  • the combination therapy can include an IL-2 agent described herein coformulated with, and/or co-administered with, one or more additional therapeutic agents, e.g., one or more additional therapeutic agents described herein.
  • the IL-2 agents are administered in combination with other therapeutic treatment modalities, e.g, other therapeutic treatment modalities described herein.
  • Such combination therapies may advantageously utilize lower dosages of the administered therapeutic agents, thus avoiding possible toxicides or complications associated with the various monotherapies.
  • Administered “in combination,” as used herein, means that two (or more) different treatments are delivered to the subject before, or during the course of the subject's affliction with a disorder.
  • two or more treatments are delivered prophy lactically, e.g., before the subject has the disorder or is diagnosed with the disorder.
  • the two or more treatments are delivered after the subject has developed or diagnosed with the disorder.
  • the delivery of one treatment is still occurring when the delivery of the second begins, so that there is overlap. This is sometimes referred to herein as “simultaneous” or “concurrent delivery.”
  • the delivery of one treatment ends before the delivery of the other treatment begins. In an embodiment of either case, the treatment is more effective because of combined administration.
  • the second treatment is more effective, e.g, an equivalent effect is seen with less of the second treatment, or the second treatment reduces symptoms to a greater extent, than would be seen if the second treatment were administered in the absence of the first treatment, or the analogous situation is seen with the first treatment.
  • delivery is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with one treatment delivered in the absence of the other.
  • the effect of the two treatments can be partially additive, wholly additive, or greater than additive.
  • the delivery can be such that an effect of the first treatment delivered is still detectable when the second is delivered.
  • the IL-2 agent is administered in combination with a second therapy (e.g., an additional agent) to treat or prevent a disorder described herein.
  • the additional agent is a second IL-2 agent, e.g, an IL-2 agent different from a first IL-2 agent.
  • Exemplary IL-2 agents that can be used in combination include, but are not limited to, any combination of the IL-2 agents described herein.
  • the additional agent is other than an IL-2 agent.
  • the additional agent can be a small molecule or a nucleic acid molecule.
  • the second therapy is a transplant, e.g., a heart transplant.
  • the second therapy comprises an immunosuppressive agent.
  • the immunosuppressive agent comprises rapamycin.
  • a method of treating a transplant rejection or a symptom thereof comprising administering to a subject in need thereof an effective amount of an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, thereby treating the transplant rejection or the symptom thereof.
  • an IL-2 agent described herein e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein
  • a method of preventing a transplant rejection or a symptom thereof comprising administering to a subject in need thereof an effective amount of an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, thereby preventing the transplant rejection or the symptom thereof.
  • an IL-2 agent described herein e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein
  • a method of modulating (e.g., increasing or inducing) immunosuppression for a transplant comprising administering to a subject in need thereof an effective amount of an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, thereby modulating (e.g., increasing or inducing) immunosuppression for a transplant.
  • an IL-2 agent described herein e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein
  • a method of conditioning a subject prior to a transplant comprising administering to a subject in need thereof an effective amount of an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, thereby conditioning the subject prior to the transplant.
  • an IL-2 agent described herein e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, thereby conditioning the subject prior to the transplant.
  • a method of selectively increasing Tregs comprising administering to a subject in need thereof an effective amount of an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, wherein the subject has received, is receiving, or will receive a transplant, thereby selectively increasing Tregs.
  • an IL-2 agent described herein e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein
  • transplant is a heart transplant.
  • a method of treating a heart disease or a symptom thereof comprising: administering to a subject in need thereof an effective amount of an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein; and providing a heart transplant to the subject, thereby treating the heart disease or the symptom thereof.
  • an IL-2 agent described herein e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein
  • IL-2 fusion protein comprises an IL-2 variant comprising:
  • the IL-2 variant further comprises the amino acid substitution T3A.
  • the IL-2 variant comprises the amino acid sequence of any of SEQ ID NOs: 4, 5, 11, 1000, 1001, or 1002, an amino acid sequence that is at least 95% identical thereto or differs by no more than 1, 2, 3, 4, or 5 amino acids therefrom, or a functional fragment thereof.
  • the Fc region comprises an Fc region of IgGl allotype m3 comprising an N297G substitution according to EU numbering.
  • Fc region comprises the amino acid sequence of SEQ ID NO: 1003, or an amino acid sequence that is at least 95% identical thereto or differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids therefrom, or a functional fragment thereof.
  • fusion protein comprises an amino acid sequence of any of SEQ ID NOs: 1004, 1005, 1006, 1007, 1008, or 1009, an amino acid sequence that is at least 95% identical thereto or differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids therefrom, or a functional fragment thereof.
  • a combination comprising an IL-2 agent described herein, e.g., an IL-2 variant described herein, or an IL-2 fusion protein described herein, and a transplant.
  • embodiment 27 or 28 further comprising one or more additional immunosuppressive agents, e.g., rapamycin.
  • additional immunosuppressive agents e.g., rapamycin.
  • transplant rejection is a heart transplant rejection.
  • IL-2 agent is administered to the subject prior to, during, and/or after the transplant.
  • Example 1 IL-2 Mutein Therapy Induces FOXP3+ Treg Expansion and Donor-Specific Allograft Tolerance
  • IL-2m an exemplary IL-2 mutein described herein induced Stat5b phosphorylation and promoted inducible Treg development in vitro by 5-fold (FIG. 1A, IB, 2A, and 2B), and by 8-fold when used in conjunction with a Cdk8/19i.
  • the IL-2 mutein expanded the Foxp3+ Treg population by 4-5-fold (FIG. 3).
  • IL-2 mutein therapy was therefore tested in a fully MHC-mismatched cardiac allograft model (BALB/c->C57BL/6) in which recipients also received low-dose rapamycin (RPM, 0.5 mg/kg/d via Alzet pump for 2 wk post-Tx).
  • RPM low-dose rapamycin
  • IL-2C exemplary IL-2 mutein
  • IL-2C IL-2 mutein

Abstract

L'invention concerne des procédés d'utilisation de protéines de fusion IL-2 pour traiter et/ou prévenir un rejet de greffe tel que le rejet de greffe cardiaque.
PCT/US2022/080728 2021-12-01 2022-12-01 Procédés d'utilisation d'agents d'interleukine-2 WO2023102463A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202163284978P 2021-12-01 2021-12-01
US63/284,978 2021-12-01
US202263348201P 2022-06-02 2022-06-02
US63/348,201 2022-06-02

Publications (1)

Publication Number Publication Date
WO2023102463A1 true WO2023102463A1 (fr) 2023-06-08

Family

ID=84981477

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/080728 WO2023102463A1 (fr) 2021-12-01 2022-12-01 Procédés d'utilisation d'agents d'interleukine-2

Country Status (3)

Country Link
US (1) US20240041981A1 (fr)
TW (1) TW202333772A (fr)
WO (1) WO2023102463A1 (fr)

Citations (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0125023A1 (fr) 1983-04-08 1984-11-14 Genentech, Inc. Préparations d'immunoglobuline recombinante, méthodes pour leur préparation, séquences d'ADN, vecteurs d'expression et cellules d'hôtes recombinantes
EP0171496A2 (fr) 1984-08-15 1986-02-19 Research Development Corporation of Japan Procédé pour la production d'un anticorps monoclonal chimérique
EP0173494A2 (fr) 1984-08-27 1986-03-05 The Board Of Trustees Of The Leland Stanford Junior University Récepteurs chimériques par liaison et expression de l'ADN
WO1986001533A1 (fr) 1984-09-03 1986-03-13 Celltech Limited Production d'anticorps chimeriques
EP0184187A2 (fr) 1984-12-04 1986-06-11 Teijin Limited Chaîne lourde d'immunoglobuline chimère souris-humaine et chimère de l'ADN codant celle-ci
GB2188638A (en) 1986-03-27 1987-10-07 Gregory Paul Winter Chimeric antibodies
WO1990002809A1 (fr) 1988-09-02 1990-03-22 Protein Engineering Corporation Production et selection de proteines de liaison diversifiees de recombinaison
EP0388151A1 (fr) 1989-03-13 1990-09-19 Celltech Limited Anticorps modifiés
WO1991000906A1 (fr) 1989-07-12 1991-01-24 Genetics Institute, Inc. Animaux chimeriques et transgeniques pouvant produire des anticorps humains
WO1991010741A1 (fr) 1990-01-12 1991-07-25 Cell Genesys, Inc. Generation d'anticorps xenogeniques
WO1991017271A1 (fr) 1990-05-01 1991-11-14 Affymax Technologies N.V. Procedes de triage de banques d'adn recombine
WO1992001047A1 (fr) 1990-07-10 1992-01-23 Cambridge Antibody Technology Limited Procede de production de chainon de paires a liaison specifique
WO1992003917A1 (fr) 1990-08-29 1992-03-19 Genpharm International Recombinaison homologue dans des cellules de mammiferes
WO1992003918A1 (fr) 1990-08-29 1992-03-19 Genpharm International, Inc. Animaux non humains transgeniques capables de produire des anticorps heterologues
WO1992009690A2 (fr) 1990-12-03 1992-06-11 Genentech, Inc. Methode d'enrichissement pour des variantes de l'hormone de croissance avec des proprietes de liaison modifiees
WO1992015679A1 (fr) 1991-03-01 1992-09-17 Protein Engineering Corporation Phage de visualisation d'un determinant antigenique ameliore
WO1992018619A1 (fr) 1991-04-10 1992-10-29 The Scripps Research Institute Banques de recepteurs heterodimeres utilisant des phagemides
WO1992020791A1 (fr) 1990-07-10 1992-11-26 Cambridge Antibody Technology Limited Methode de production de chainons de paires de liaison specifique
EP0519596A1 (fr) 1991-05-17 1992-12-23 Merck & Co. Inc. Procédé pour réduire l'immunogénécité des domaines variables d'anticorps
WO1993001288A1 (fr) 1991-07-08 1993-01-21 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Phagemide utile pour trier des anticorps
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
WO1994004678A1 (fr) 1992-08-21 1994-03-03 Casterman Cecile Immunoglobulines exemptes de chaines legeres
US5585089A (en) 1988-12-28 1996-12-17 Protein Design Labs, Inc. Humanized immunoglobulins
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US8602269B2 (en) 2009-09-14 2013-12-10 Guala Dispensing S.P.A. Trigger sprayer
US20140286898A1 (en) * 2013-03-14 2014-09-25 Amgen Inc. Interleukin-2 muteins for the expansion of t-regulatory cells
WO2016164937A2 (fr) 2015-04-10 2016-10-13 Amgen Inc. Mutéines de l'interleukine-2 pour l'expansion de lymphocytes t régulateurs
AU2016246152A1 (en) * 2015-04-10 2017-11-02 Amgen Inc. Interleukin-2 muteins for the expansion of T-regulatory cells
US20180037634A1 (en) 2016-08-02 2018-02-08 Visterra, Inc. Engineered polypeptides and uses thereof
WO2019010224A1 (fr) * 2017-07-03 2019-01-10 Torque Therapeutics, Inc. Molécules de fusion ciblant des cellules régulatrices immunitaires et leurs utilisations
US20210024601A1 (en) * 2019-07-26 2021-01-28 Visterra, Inc. Interleukin-2 agents and uses thereof

Patent Citations (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0125023A1 (fr) 1983-04-08 1984-11-14 Genentech, Inc. Préparations d'immunoglobuline recombinante, méthodes pour leur préparation, séquences d'ADN, vecteurs d'expression et cellules d'hôtes recombinantes
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
EP0171496A2 (fr) 1984-08-15 1986-02-19 Research Development Corporation of Japan Procédé pour la production d'un anticorps monoclonal chimérique
EP0173494A2 (fr) 1984-08-27 1986-03-05 The Board Of Trustees Of The Leland Stanford Junior University Récepteurs chimériques par liaison et expression de l'ADN
WO1986001533A1 (fr) 1984-09-03 1986-03-13 Celltech Limited Production d'anticorps chimeriques
EP0184187A2 (fr) 1984-12-04 1986-06-11 Teijin Limited Chaîne lourde d'immunoglobuline chimère souris-humaine et chimère de l'ADN codant celle-ci
GB2188638A (en) 1986-03-27 1987-10-07 Gregory Paul Winter Chimeric antibodies
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US5648260A (en) 1987-03-18 1997-07-15 Scotgen Biopharmaceuticals Incorporated DNA encoding antibodies with altered effector functions
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
WO1990002809A1 (fr) 1988-09-02 1990-03-22 Protein Engineering Corporation Production et selection de proteines de liaison diversifiees de recombinaison
US5693762A (en) 1988-12-28 1997-12-02 Protein Design Labs, Inc. Humanized immunoglobulins
US5693761A (en) 1988-12-28 1997-12-02 Protein Design Labs, Inc. Polynucleotides encoding improved humanized immunoglobulins
US5585089A (en) 1988-12-28 1996-12-17 Protein Design Labs, Inc. Humanized immunoglobulins
EP0388151A1 (fr) 1989-03-13 1990-09-19 Celltech Limited Anticorps modifiés
WO1991000906A1 (fr) 1989-07-12 1991-01-24 Genetics Institute, Inc. Animaux chimeriques et transgeniques pouvant produire des anticorps humains
WO1991010741A1 (fr) 1990-01-12 1991-07-25 Cell Genesys, Inc. Generation d'anticorps xenogeniques
WO1991017271A1 (fr) 1990-05-01 1991-11-14 Affymax Technologies N.V. Procedes de triage de banques d'adn recombine
WO1992020791A1 (fr) 1990-07-10 1992-11-26 Cambridge Antibody Technology Limited Methode de production de chainons de paires de liaison specifique
WO1992001047A1 (fr) 1990-07-10 1992-01-23 Cambridge Antibody Technology Limited Procede de production de chainon de paires a liaison specifique
WO1992003918A1 (fr) 1990-08-29 1992-03-19 Genpharm International, Inc. Animaux non humains transgeniques capables de produire des anticorps heterologues
WO1992003917A1 (fr) 1990-08-29 1992-03-19 Genpharm International Recombinaison homologue dans des cellules de mammiferes
WO1992009690A2 (fr) 1990-12-03 1992-06-11 Genentech, Inc. Methode d'enrichissement pour des variantes de l'hormone de croissance avec des proprietes de liaison modifiees
WO1992015679A1 (fr) 1991-03-01 1992-09-17 Protein Engineering Corporation Phage de visualisation d'un determinant antigenique ameliore
WO1992018619A1 (fr) 1991-04-10 1992-10-29 The Scripps Research Institute Banques de recepteurs heterodimeres utilisant des phagemides
EP0519596A1 (fr) 1991-05-17 1992-12-23 Merck & Co. Inc. Procédé pour réduire l'immunogénécité des domaines variables d'anticorps
WO1993001288A1 (fr) 1991-07-08 1993-01-21 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Phagemide utile pour trier des anticorps
WO1994004678A1 (fr) 1992-08-21 1994-03-03 Casterman Cecile Immunoglobulines exemptes de chaines legeres
US8602269B2 (en) 2009-09-14 2013-12-10 Guala Dispensing S.P.A. Trigger sprayer
US20140286898A1 (en) * 2013-03-14 2014-09-25 Amgen Inc. Interleukin-2 muteins for the expansion of t-regulatory cells
WO2016164937A2 (fr) 2015-04-10 2016-10-13 Amgen Inc. Mutéines de l'interleukine-2 pour l'expansion de lymphocytes t régulateurs
AU2016246152A1 (en) * 2015-04-10 2017-11-02 Amgen Inc. Interleukin-2 muteins for the expansion of T-regulatory cells
US20180037634A1 (en) 2016-08-02 2018-02-08 Visterra, Inc. Engineered polypeptides and uses thereof
WO2018052556A1 (fr) 2016-08-02 2018-03-22 Visterra, Inc. Polypeptides synthétiques et leurs utilisations
WO2019010224A1 (fr) * 2017-07-03 2019-01-10 Torque Therapeutics, Inc. Molécules de fusion ciblant des cellules régulatrices immunitaires et leurs utilisations
US20210024601A1 (en) * 2019-07-26 2021-01-28 Visterra, Inc. Interleukin-2 agents and uses thereof
WO2021021606A1 (fr) 2019-07-26 2021-02-04 Visterra, Inc. Agents d'interleukine 2 et utilisations de ceux-ci

Non-Patent Citations (52)

* Cited by examiner, † Cited by third party
Title
"Antibody Engineering Lab Manual", SPRINGER-VERLAG, article "Protein Sequence and Structure Analysis of Antibody Variable Domains"
"Sustained and Controlled Release Drug Delivery Systems", 1978, MARCEL DEKKER, INC.
"Uniprot", Database accession no. P60568
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 10
ALTSCHUL ET AL., NUCLEIC ACIDS RES., vol. 25, 1997, pages 3389 - 3402
BARBAS ET AL., PNAS, vol. 88, 1991, pages 7978 - 7982
BEIDLER ET AL., J. IMMUNOL., vol. 141, 1988, pages 4053 - 4060
BOOTH ET AL., MABS., vol. 10, no. 7, 2018, pages 1098 - 1110
BRUGGEMAN ET AL., EURJ IMMUNOL, vol. 21, 1991, pages 1323 - 1326
BRUGGEMAN ET AL., YEAR IMMUNOL, vol. 7, 1993, pages 33 - 40
CHOTHIA, C. ET AL., J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
CLACKSON ET AL., NATURE, vol. 352, 1991, pages 624 - 628
COLCHER, D. ET AL., ANN N YACΑD SCI, vol. 880, 1999, pages 263 - 80
E. MEYERSW. MILLER, CABIOS, vol. 4, 1989, pages 11 - 17
FUCHS ET AL., BIO/TECHNOLOGY, vol. 9, 1991, pages 1370 - 1372
GARRAD ET AL., BIOLTECHNOLOGY, vol. 9, 1991, pages 1373 - 1377
GRAM ET AL., PNAS, vol. 89, 1992, pages 3576 - 3580
GREEN, L.L. ET AL., NATURE GENET., vol. 7, 1994, pages 13 - 21
GRIFFTHS ET AL., EMBO J, vol. 12, 1993, pages 725 - 734
HAWKINS ET AL., J MOL BIOL, vol. 226, 1992, pages 889 - 896
HAY ET AL., HUMANTIBODHYBRIDOMAS, vol. 3, 1992, pages 81 - 85
HOOGENBOOM ET AL., NUC ACID RES, vol. 19, 1991, pages 4133 - 4137
HUSE ET AL., SCIENCE, vol. 246, 1989, pages 1275 - 1281
HUSTON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 5879 - 5883
J. SEHOULI ET AL., EPIGENETICS, vol. 6, no. 2, 2011, pages 236 - 246
JONES ET AL., NATURE, vol. 321, 1986, pages 552 - 525
KIKUTANI ET AL., CIBA FOUNDATION SYMPOSIUM, vol. 147, 1989, pages 23 - 31
LIAO ET AL., CURR OPIN IMMUNOL., vol. 23, no. 5, 2011, pages 598 - 604
LIU ET AL., J. IMMUNOL., vol. 139, 1987, pages 3521 - 3526
LOBUGLIO ET AL., HYBRIDOMA, vol. 5, 1986, pages 5117 - 5123
LONBERG ET AL., NATURE, vol. 314, 1985, pages 446 - 449
MALEKCASTRO, IMMUNITY, vol. 33, no. 2, 2010, pages 153 - 165
MAVERAKIS ET AL., JOURNAL OF AUTOIMMUNITY, vol. 57, no. 6, 2015, pages 1 - 13
MORRISON, S. L., SCIENCE, vol. 229, 1985, pages 1202 - 1207
MORRISON, S.L. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 81, 1994, pages 6851 - 6855
NEEDLEMANWUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 444 - 453
NISHIMURA ET AL., CANC. RES., vol. 47, 1987, pages 999 - 1005
OI ET AL., BIOTECHNIQUES, vol. 4, 1986, pages 214
PEIPP ET AL., BLOOD, vol. 112, no. 6, 2008, pages 2390 - 2399
REITER, Y., CLIN CANCER RES, vol. 2, 1996, pages 245 - 52
SALEH ET AL., CANCER IMMUNOL. IMMUNOTHER., vol. 32, 1990, pages 180 - 190
SHAW ET AL., J. NATL CANCER INST., vol. 80, 1988, pages 1553 - 1559
SHIBUYAHONDA, SPRINGER SEMINARS IN IMMUNOPATHOLOGY, vol. 28, no. 4, 2006, pages 377 - 82
STADLMANN ET AL., PROTEOMICS, vol. 8, no. 14, 2008, pages 2858 - 2871
STADLMANN, PROTEOMICS, vol. 9, no. 17, 2009, pages 4143 - 4153
SUN ET AL., PNAS, vol. 84, 1987, pages 3439 - 3443
TUAILLON ET AL., PNAS, vol. 90, 1993, pages 3720 - 3724
VERHOEYAN ET AL., SCIENCE, vol. 239, 1988, pages 1534 - 1043
VON BUBNOFF ET AL., CLINICAL AND EXPERIMENTAL DERMATOLOGY, vol. 28, no. 2, 2003, pages 184 - 7
WOZNIAK-KNOPP ET AL., PROTEIN ENG DES, vol. 23, no. 4, 2010, pages 289 - 297
XIN XIAO ZHENG ET AL: "The balance of deletion and regulation in allograft tolerance", IMMUNOLOGICAL REVIEWS, WILEY-BLACKWELL PUBLISHING, INC, US, vol. 196, no. 1, 10 November 2003 (2003-11-10), pages 75 - 84, XP071455034, ISSN: 0105-2896, DOI: 10.1046/J.1600-065X.2003.00089.X *
ZHU ET AL., JOURNAL OF IMMUNOLOGY, vol. 166, no. 5, 2001, pages 3266 - 76

Also Published As

Publication number Publication date
US20240041981A1 (en) 2024-02-08
TW202333772A (zh) 2023-09-01

Similar Documents

Publication Publication Date Title
US20220040264A1 (en) Il-15/il-15ra heterodimeric fc fusion proteins and uses thereof
EP3464367B1 (fr) Protéines de liaison bispécifiques se liant à une protéine immunomodulatrice et à un antigène tumoral
EP3504242B1 (fr) Anticorps anti-ox40 et leurs utilisations
KR101197542B1 (ko) 항-cd20 항체 및 이용 방법
US20210024601A1 (en) Interleukin-2 agents and uses thereof
KR20210013708A (ko) 항-ox40 항체 및 사용 방법
JP2022513653A (ja) 修飾された重鎖定常領域を含む抗体
JP2017536101A (ja) Ccr6と結合する抗体およびその使用
AU2022310847A1 (en) Anti-c-c motif chemokine receptor 8 (ccr8) antibodies and methods of use
CA3157042A1 (fr) Procedes de traitement du cancer utilisant des anticorps anti-ox40 en combinaison avec des anticorps anti-tigit
WO2022258673A1 (fr) Anticorps multispécifiques se liant à cd20, nkp46, cd16 et conjugués à il-2
AU2021374083A1 (en) Anti-cd19 agent and b cell targeting agent combination therapy for treating b cell malignancies
US20240041981A1 (en) Methods of using interleukin-2 agents
KR20230104611A (ko) 항-cd94 항체 및 이의 사용 방법
KR20230129018A (ko) 인터류킨-2 작용제를 사용하는 방법
JP2023503031A (ja) 化学療法剤との組合せでの抗ox40抗体による癌治療の方法
EA045547B1 (ru) Антитела к ox40 и способы применения
AU2013203712A1 (en) Binding Proteins, Including Antibodies, Antibody Derivatives And Antibody Fragments, That Specifically Bind CD154 And Uses Thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22843966

Country of ref document: EP

Kind code of ref document: A1